Sélection de la langue

Search

Sommaire du brevet 2970664 

Énoncé de désistement de responsabilité concernant l'information provenant de tiers

Une partie des informations de ce site Web a été fournie par des sources externes. Le gouvernement du Canada n'assume aucune responsabilité concernant la précision, l'actualité ou la fiabilité des informations fournies par les sources externes. Les utilisateurs qui désirent employer cette information devraient consulter directement la source des informations. Le contenu fourni par les sources externes n'est pas assujetti aux exigences sur les langues officielles, la protection des renseignements personnels et l'accessibilité.

Disponibilité de l'Abrégé et des Revendications

L'apparition de différences dans le texte et l'image des Revendications et de l'Abrégé dépend du moment auquel le document est publié. Les textes des Revendications et de l'Abrégé sont affichés :

  • lorsque la demande peut être examinée par le public;
  • lorsque le brevet est émis (délivrance).
(12) Demande de brevet: (11) CA 2970664
(54) Titre français: NOUVEAUX COMPOSES
(54) Titre anglais: NOVEL COMPOUNDS
Statut: Morte
Données bibliographiques
(51) Classification internationale des brevets (CIB):
  • C07D 403/04 (2006.01)
  • A61K 31/4155 (2006.01)
  • A61K 31/4178 (2006.01)
  • A61P 11/00 (2006.01)
  • C07D 405/04 (2006.01)
(72) Inventeurs :
  • ARMANI, ELISABETTA (Italie)
  • CAPALDI, CARMELIDA (Italie)
  • SUTTON, JONATHAN MARK (Italie)
  • HEALD, ROBERT ANDREW (Italie)
  • RIZZI, ANDREA (Italie)
(73) Titulaires :
  • CHIESI FARMACEUTICI S.P.A. (Italie)
(71) Demandeurs :
  • CHIESI FARMACEUTICI S.P.A. (Italie)
(74) Agent: BERESKIN & PARR LLP/S.E.N.C.R.L.,S.R.L.
(74) Co-agent:
(45) Délivré:
(86) Date de dépôt PCT: 2015-12-11
(87) Mise à la disponibilité du public: 2016-06-23
Licence disponible: S.O.
(25) Langue des documents déposés: Anglais

Traité de coopération en matière de brevets (PCT): Oui
(86) Numéro de la demande PCT: PCT/EP2015/079371
(87) Numéro de publication internationale PCT: WO2016/096638
(85) Entrée nationale: 2017-06-12

(30) Données de priorité de la demande:
Numéro de la demande Pays / territoire Date
14198044.1 Office Européen des Brevets (OEB) 2014-12-15

Abrégés

Abrégé français

L'invention concerne des dérivés d'imidazolone présentant des propriétés inhibitrices de l'élastase neutrophile humaine, et leur utilisation thérapeutique.


Abrégé anglais

This invention relates to imidazolone derivatives having human neutrophil elastase inhibitory properties, and their use in therapy.

Revendications

Note : Les revendications sont présentées dans la langue officielle dans laquelle elles ont été soumises.


80
CLAIMS
1. A compound of general formula (I)
Image
wherein
A is -CH or ¨N-;
B is -CH or ¨N-;
D is -CH or ¨N-;
X is -CH or ¨N-;
R3 is -CF3, CHF2 or halogen;
W is a 5 or 6-membered heteroaryl ring, optionally substituted with one or
more
substituents selected from -CN, -NO2, -O(C1-C6)alkyl, -(C1-C3)alkyl-O-(C1-
C6)alkyl, -(C1-
C3)alkylamino, -(C3-C8)cycloalkyl, -OH, (C1-C3)alkyl, halo, -CF3 and -SO2(C1-
C4)alkyl;
R1 is H or is selected in the group consisting of -C(O)R4, -(CH2)t-(CHR6)n-R4,
-
(CH2)t-(CHR6)nS(O2)-NR4Rs, -S(O2)-R4, -(CH2)t-(CHR6)n-C(O)-NR4Rs, -(CH2)t-
(CHR6)n-
C(O)O-R4;
R4 is H or is selected in the group consisting of -(C1-C6)alkyl, -(C2-C6)alkyl-

NR7R8, -(C2-C6)alkyl-N+R7R8R9, -(C2-C6)alkyl-O-(C1-C4)alkyl, -(C2-C6)alkyl-
S(O2)(C1-
C4)alkyl, -(CH2)t-aryl, -(CH2)t-(C3-C8)cycloalkyl, -(CH2)t-(C3-
C8)heterocycloalkyl and -
(CH2)t-heteroaryl, optionally substituted with one or more substituents
selected from -CN,
-NO2, -O(C1-C6)alkyl, -(C1-
C3)alkyl-O-(C1-C6)alkyl,
-(C1-C3)alkylamino, -(C3-C8)cycloalkyl, -OH, (C1-C3)alkyl, halo, -CF3 and
¨SO2(C1-
C4)alkyl;

81
n is 0 or an integer selected from 1, 2 or 3;
t is 0 or an integer selected from 1, 2 or 3;
R5 is H or -(C1-C6)alkyl;
R6 is H, -(C1-C6)alkyl, -CF3 or -CHF2;
R7 is H or -(C1-C6)alkyl;
R8 is H or -(C1-C6)alkyl;
R9 is H or -(C1-C6)alkyl;
wherein alternatively R4 and R5 together with the nitrogen atom they are
linked to,
may form a (C3-C8)heterocycloalkyl or heteroaryl ring system, optionally
substituted with
one or more substituents selected from -CN, -NO2, -O(C1-C6)alkyl, -(C1-
C3)alky1-O-(C1-
C6)alkyl, -(C1-C3)alkylamino, -(C3-C8)cycloalkyl, -OH, (C1-C3)alkyl, halo, -
CF3 and ¨
SO2(C1-C4)alkyl, wherein the heteroatoms of the (C3-C8)heterocycloalkyl or
heteroaryl ring
system are maximum three and selected from -N, -NH, -O, -S- and -SO2;
R2 is H or is selected from the group consisting of, -SMe, ¨S(O)R7, ¨S(O2)R7
and
halogen; and pharmaceutically acceptable salts thereof
2. A compound according to claim 1, wherein A, B, D and X are ¨CH.
3. A compound according to any preceding claim, wherein W is selected from
the
group consisting of pyrazolyl, imidazolyl and thiazolyl.
4. A compound according to any preceding claim, wherein R3 is -CF3.
5. A compound according to any preceding claim, wherein R2 is H or is
selected from
the group consisting of halogen and ¨S(O2)R7 wherein R7 is -(C1-C6)alkyl.
6. A compound according to any preceding claim, wherein, R1 is -(CH2)t-
(CHR6)n-
C(O)-NR4R5 wherein R4 is H or is selected in the group consisting of -(C1-
C6)alkyl,
optionally substituted with one or more groups selected from -O(C1-C6)alkyl,
-(C1-C3)alkylamino, ¨SO2(C1-C4)alkyl and -OH or R4 is selected in the group
consisting of ¨
(CH2)t-(C3-C8)cycloalkyl, ¨(CH2)t-(C3-C8)heterocycloalkyl, -(C2-C6)alkyl-
NR7R8, -(CH2)t-
aryl, -(CH2)t-heteroaryl and -(C2-C6)alkyl-N+R7R8R9, optionally substituted
with one or
more -(C1-C6)alkyl or ¨SO2(C1-C4)alkyl; R5 is H or -(C1-C6)alkyl; or wherein
alternatively

82
R4 and Rs together with the nitrogen atom they are linked to, may form a (C3-
C8)heterocycloalkyl or heteroaryl ring system, optionally substituted with one
or more
substituents selected from ¨OH and (C1-C3)alkyl, wherein the heteroatoms of
the (C3-
C8)heterocycloalkyl or heteroaryl ring system are maximum three and selected
from -N, -
NH, -O, -S- and -SO2; R6 is H, -(C1-C6)alkyl, -CF3 or -CHF2; R7 is H or -(C1-
C6)alkyl; R8
is H or -(C1-C6)alkyl; t is 0 or an integer selected from 1, 2 or 3; n is 0 or
an integer selected
from 1, 2 or 3.
7. A compound according to claims 1-5, wherein R1 is -(CH2)t-(CHR6)n-C(O)O-
R4
wherein R4 is H or -(C1-C6)alkyl, optionally substituted with one or more
substituents
selected from -CN, -NO2, -O(C1-C6)alkyl, -OH, (C1-C3)alkyl, halo and -CF3; R6
is H,
-(C1-C6)alkyl, -CF3 or -CHF2; t is 0 or an integer selected from 1, 2 or 3; n
is 0 or an integer
selected from 1, 2 or 3.
8. A compound according to claims 1-5, wherein R1 is -(CH2)t-(CHR6)nS(O2)-
NR4R5
wherein R4 is H or is selected in the group consisting of -(C1-C6)alkyl and -
(C3-
C8)cycloalkyl; R5 is H or -(C1-C6)alkyl; t is 0 or an integer selected from 1,
2 or 3; n is 0
or an integer selected from 1, 2 or 3.
9. A compound according to claims 1-5, wherein R1 is -S(O2)-R4, R4 is H or
is selected
in the group consisting of ¨(CH2)t-(C3-C8)cycloalkyl and -(C1-C6)alkyl; t is 0
or an integer
selected from 1, 2 or 3.
10. A compound as claimed in any of the preceding claims, which is selected
in the
group consisting of
4-(5-{5-Methyl-2-oxo-1-[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazol-4-
yl} -
1H-pyrazol-1-yl)benzonitrile;
4-(5-{5-Methyl-3-[3-(methylsulfonyl)propyl]-2-oxo-1-[3-
(trifluoromethyl)phenyl]-2,3 -
dihydro-1H-imidazol-4-yl)-1H-pyrazol-1-yl)benzonitrile;
4-(5-{3,5-Dimethyl-2-oxo-1-[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazol-
4-
yl}-1H-pyrazol-1-yl)benzonitrile ;
Ethyl-5-[1-(4-cyanophenyl)-1H-pyrazol-5-yl]-4-methyl-2-oxo-3-[3-

83
(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxylate;
4-(5-{3-Acetyl-5-methyl-2-oxo-1-[3(trifluoromethyl)phenyl]-2,3-dihydro-1H-
imidazol-4-yl}-1H-pyrazol-1-yl)benzonitrile;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-N-ethyl-4-methyl-2-oxo-3-[3-
(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-N-(2-hydroxyethyl)-4-methyl-2-oxo-3-[3-
(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-N-(3-hydroxypropyl)-4-methyl-2-oxo-3-[3-

(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-N-[3-(dimethylamino)propyl]-4-methyl-2-
oxo-3-[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
4-(5-{3-Butanoyl-5-methyl-2-oxo-1-[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-
imidazol-4-yl}-1H-pyrazol-1-yl)benzonitrile;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-N-(2-methoxyethyl)-4-methyl-2-oxo-3-[3-
(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-N-[2-(dimethylamino)ethyl]-4-methyl-2-
oxo-
3-[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-4-methyl-N-[2-(methylsulfonyl)ethyl]-2-
oxo-
3-[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-4-methyl-2-oxo-N-(tetrahydrofuran-3-yl)-
3-
[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-4-methyl-2-oxo-N-[(3S)-tetrahydrofuran-3-
yl]-
3-[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-4-methyl-2-oxo-N-[(3R)-tetrahydrofuran-3-

yl]-3-[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-carboxamide;
5-[1-(4-Cyanophenyl)-1H-pyrazol-5-yl]-4-methyl-N-[(1-methyl-1H-pyrazol-3-
yl)methyl]-2-oxo-3-[3-(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazole-1-
carboxamide;

84
Methy1-5 41 -(4-cyanopheny1)-1H-pyrazol-5-yl] -4-methy1-2-oxo-3 - [3 -
(trifluoromethyl)phenyl] -2,3 -dihydro-1H-imidazole- 1 -carboxylate;
4-(5-15-Methy1-2-oxo-3 -pentanoyl- 1 43 -(trifluoromethyl)phenyl] -2,3 -
dihydro- 1 H-
imidazol-4-yll - 1 H-pyrazol- 1 -yObenzonitrile;
tert-Butyl- { 5 41 -(4-cyanopheny1)-1H-pyrazol-5-y1]-4-methy1-2-oxo-3 - [3 -
(trifluoromethyl)pheny1]-2,3 -dihydro- 1 H-imidazol- 1 -y1} acetate;
5- [1 -(4-Cyanopheny1)- 1 H-pyrazol-5-y1]-N-ethyl-N,4-dimethy1-2-oxo-343-
(trifluoromethyl)phenyl]-2,3 -dihydro- 1 H-imidazole- 1 -carboxamide;
{5 - [1 -(4-Cyanopheny1)- 1 H-pyrazol-5-y1]-4-methy1-2-oxo-3 - [3-
(trifluoromethyl)phenyl] -2,3 -dihydro- 1 H-imidazol- 1 -y1} acetic acid;
2- { 5- [1 -(4-Cyanopheny1)- 1 H-pyrazol-5-yl] -4-methy1-2-oxo-3 - [3 -
(trifluoromethyl)phenyl] -2,3 -dihydro- 1H-imidazol- 1 -y1} -N-
methylacetamide;
Benzenesulfonate 3 - { 5 41 -(4-cyanopheny1)-1H-pyrazol-5-yl] -4-methy1-2-oxo-
3 - [3 -
(trifluoromethyl)pheny1]-2,3 -dihydro- 1 H-imidazol-1 -y1} carbonypamino] -
N,N,N-
trimethylpropan- 1 -aminium;
Benzenesulfonate 2- [( { 5- [1 -(4-cyanopheny1)- 1 H-pyrazol-5-yl] -4-methy1-2-
oxo-3 43 -
(trifluoromethyl)phenyll -2,3-dihydro- 1H-imidazol- 1 -yll carbonyl)amino]-
N,N,N-
trimethylethanaminium;
445- { 3 -Buty1-5-methy1-2-oxo-1- [3 -(trifluoromethyl)pheny1]-2,3 -dihydro- 1
H-imidazol-
4-y11 - 1 H-pyrazol- 1 -yl)benzonitrile;
4-(5- 5-Methy1-3 -(methylsulfony1)-2-oxo-1 43 -(trifluoromethyl)phenyl] -2,3 -
dihydro-
111-imidazol-4-yll - 1 H-pyrazol- 1 -yObenzonitrile;
4- { 5- [5-Methy1-3 -(1 -methyl-buty1)-2-oxo- 1 -(3 -trifluoromethyl-pheny1)-
2,3 -dihydro-
1H-imidazol-4-yl]pyrazol- 1 -y1} -benzonitrile;
4- { 5- [3 -(2-Dimethylamino-ethyl)-5-methy1-2-oxo-1 -(3 -trifluoromethyl-
pheny1)-2,3 -
dihydro- 1 H-imidazol-4-yl] -pyrazol- 1 -yll -benzonitrile;
4- { 543 -(3 -Dimethylamino-propy1)-5-methy1-2-oxo- 1 -(3 -trifluoromethyl-
pheny1)-2,3 -
dihydro- 1 H-imidazol-4-yl] -pyrazol-1 -y1} -benzonitrile;

85
4- { 5- [5-Methyl-2-oxo-3-(propane-1 -sulfonyl)-1 -(3 -trifluoromethyl-phenyl)-
2,3-
dihydro-1H-imidazol-4-yl}-pyrazol-1-yl}-benzonitrile;
4- {5 - [3 -Cyclopropanesulfonyl-5 -methyl-2-oxo- 1 -(3 -trifluoromethyl-
phenyl)-2,3 -
dihydro-1H-imidazol-4-yl]-pyrazol-1-yl}-benzonitrile;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-sulfonic acid ethylamide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-sulfonic acid cyclopentylamide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-sulfonic acid cyclopropylamide;
4-{5-[5-Methyl-2-oxo-3-(2-oxo-2-pyrrolidin-1-yl-ethyl)-1-(3-trifluoromethyl-
phenyl)-
2,3 -dihydro-1H-imidazol-4-yl] -pyrazol- 1 -yl -benzonitrile;
2-[5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-2,3-dihydro-imidazol-1-yl]-N,N-dimethyl-acetamide;
4- { 5 -[5 -Methyl-3 - [2-(4-methyl-piperazin- 1 -yl)-2-oxo-ethyl]-2-oxo-1 -(3
-
trifluoromethyl-phenyl)-2,3 -dihydro-1H-imidazol-4-yl] -pyrazol- 1 -yl -
benzonitrile;
2-[5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-2,3-dihydro-imidazol-1-yl]-N-(3-dimethylamino-propyl)-acetamide
formic
acid salt;
2-[5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-2,3-dihydro-imidazol-1-yl]-N-cyclobutyl-N-methyl-acetamide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-carboxylic acid cyclobutylamide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-carboxylic acid cyclopropylamide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-carboxylic acid (1-ethanesulfonyl-piperidin-4-yl)-
amide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-

86
2,3-dihydro-imidazole-1-carboxylic acid (tetrahydro-pyran-4-yl)-amide;
5- [2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-carboxylic acid cyclopentylamide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole- 1-carboxylic acid ((R)-1-methyl-pyrrolidin-3-yl)-amide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole- 1-carboxylic acid ((S)- 1 -methyl-pyrrolidin-3-yl)-
amide;
Mixture of 5- [2-(4-Cyano-phenyl)-2H-pyrazol-3 -yl] -4-methyl-2-oxo-3-
(3-
trifluoromethyl-phenyl)-2,3-dihydro-imidazole-1-carboxylic acid ((3R,4S)-4-
hydroxy-
tetrahydro-furan-3-yl)-amide and 5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-
methyl-2-
oxo-3-(3-trifluoromethyl-phenyl)-2,3-dihydro-imidazole-1-carboxylic acid
((3S,4R)-4-
hydroxy-tetrahydro-furan-3-yl)-amide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3 -dihydro-imidazole- 1-carboxylic acid (pyridin-2-ylmethyl)-amide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-carboxylic acid (pyridin-3-ylmethyl)-amide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-carboxylic acid (pyridin-4-ylmethyl)-amide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3 -(3-trifluoromethyl-
phenyl)-
2,3 -dihydro-imidazole-1-carboxylic acid (pyrazin-2-ylmethyl)-amide;
5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-carboxylic acid ((1R,2R)-2-hydroxy-cyclopentyl)-amide;

5-[2-(4-Cyano-phenyl)-2H-pyrazol-3-yl] -4-methyl-2-oxo-3 -(3 -trifluoromethyl-
phenyl)-
2,3-dihydro-imidazole-1-carboxylic acid (1-methyl-piperidin-4-ylmethyl)-amide;

3-Methanesulfonyl-4-{ 5-[5-methyl-2-oxo-1-(3-trifluoromethyl-phenyl)-2,3-
dihydro-
1H-imidazol-4-yl]-pyrazol-1-yl) -benzonitrile;
5- [2-(2-Bromo-4-cyano-phenyl)-2H-pyrazol-3-yl]-4-methyl-2-oxo-3-(3-
trifluoromethyl-phenyl)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide;

87

5-[1 -(2-Bromo-4-cyano-phenyl)-1H-pyrazol-3 -yl] -4-methyl-2-oxo-3 -(3 -
trifluoromethyl-phenyl)-2,3 -dihydro-imidazole-1 -carboxylic acid
cyclopentylamide;
5- [2 -(4-Cyano -2-fluoro-phenyl)-2H-pyrazol-3-yl] -4-methyl-2 -oxo-3 -(3-
trifluoromethyl-phenyl)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide;
4- [5'-Methyl-2'-oxo-1 '-(3 -trifluoromethyl-phenyl)-2',3 '-dihydro -1 'H-
[2,4'] biimidazolyl-
1 -yl]-benzonitrile ;
1-(4-Cyano-phenyl)-5 '-methyl-2'-oxo -1'-(3 -trifluoromethyl-phenyl)-1 ',2'-
dihydro -1H-
[2,41 biimidazolyl-3 '-carboxylic acid cyclopentylamide;
1 -(4-Cyano -phenyl)-5 '-methyl-2'-oxo-1 '-(3 -trifluoromethyl-phenyl)-1 ',2'-
dihydro -1H-
[2,4']biimidazolyl-3'-carboxylic acid cyclobutylamide;
1-(4-Cyano-phenyl)-5'-methyl-2'-oxo -1 '-(3 -trifluoromethyl-phenyl)-1 ',2'-
dihydro -1H-
[2,4']biimidazolyl-3 '-carboxylic acid 4-methanesulfonyl-benzylamide;
5-[2-(4-Cyano -phenyl)-2 H-[1,2,4] triazol-3 -yl] -4-methyl-2 -oxo -3 -(3 -
trifluoromethyl-
phenyl)-2,3-dihydro-imidazole-1-carboxylic acid cyclopentylamide;
and pharmaceutically acceptable salts thereof.
11. A pharmaceutical composition comprising a compound as claimed in any of
the
preceding claims together with one or more pharmaceutically acceptable
carriers or
excipients.
12. A pharmaceutical composition as claimed in claim 11, which is adapted
for oral
administration or administration by the pulmonary route.
13. A compound as claimed in any of claims 1 to 10 for use in the treatment
of, or for
use in the manufacture of a medicament for use in the treatment of a disease
or condition
in which HNE is implicated.
14. A method of treatment of a disease or condition in which HNE is
implicated,
comprising administering to a subject suffering from such disease an effective
amount of a
compound as claimed in any of claims 1 to 10.
15. A compound for use according to claim 13 or a method of treatment
according to
claim 14, wherein the disease or condition is COPD, bronchiectasis, chronic
bronchitis,


88

lung fibrosis, pneumonia, ARDS, pulmonary emphysema, smoking-induced emphysema

or cystic fibrosis, asthma, rhinitis, psoriasis, atopic dermatitis, non-atopic
dermatitis,
Crohn's disease, ulcerative colitis, or irritable bowel disease.

Description

Note : Les descriptions sont présentées dans la langue officielle dans laquelle elles ont été soumises.


CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
1
NOVEL COMPOUNDS
Field of the Invention
This invention relates to imidazolone derivatives having human neutrophil
elastase
inhibitory properties, and their use in therapy.
Background to the invention
Human neutrophil elastase (HNE) is a 32 kDa serine proteinase found in the
azurophilic granules of neutrophils. It has a role in the degradation of a
wide range of
extracellular matrix proteins, including fibronectin, laminin, proteoglycans,
Type III and
Type IV collagens as well as elastin (Bieth, G. In Regulation of Matrix
accumulation,
Mecham, R.P. (Eds), Academic Press, NY, USA 1986, 217-306). HNE has been for
long
considered to play an important role in homeostasis through repair and
disposal of damaged
tissues via degradation of the tissue structural proteins. It is also relevant
in the defense
against bacterial invasion by means of degradation of the bacterial body. In
addition to its
effects on matrix tissues, HNE has been implicated in the upregulation of IL-8
gene
expression and also induces IL-8 release from the epithelial cells of the
lung. In animal
models of Chronic Obstructive Pulmonary Disease (COPD) induced by tobacco
smoke
exposure both small molecule inhibitors and protein inhibitors of HNE inhibit
the
inflammatory response and the development of emphysema (Wright, J.L. et al.
Am.
I Respir. Grit. Care Med. 2002, 166, 954-960; Churg, A. et al. Am. I Respir.
Crit. Care
Med. 2003, 168, 199-207). Thus, HNE may play a role both in matrix destruction
and in
amplifying inflammatory responses in chronic respiratory diseases where
neutrophil influx
is a characteristic feature. Indeed, HNE is believed to play a role in several
pulmonary
diseases, including COPD, cystic fibrosis (CF), acute respiratory distress
syndrome
(ARDS), pulmonary emphysema, pneumonia and lung fibrosis. It is also
implicated in
several cardiovascular diseases in which tissue remodelling is involved, for
example, in
heart failure and the generation of ischemic tissue injury following acute
myocardial
infarction.

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
2
COPD is an umbrella term encompassing three different pathological conditions,
all
of which contribute to limitation of airflow: chronic bronchitis, emphysema
and small-
airway disease. Generally all three will exist to varying extents in patients
presenting with
COPD, and all three may be due to neutrophil-mediated inflammation, as
supported by the
increased number of neutrophils observed in bronchoalveolar leakage (BAL)
fluids of
COPD patients (Thompson, A.B.; Daughton, D.; et al. Am. Rev. Respir. Dis.
1989, 140,
1527-1537). The major pathogenic determinant in COPD has long been considered
to be
the protease-anti-protease balance (also known as the "elastase:anti-elastase
hypothesis"),
in which an imbalance of HNE and endogenous antiproteases such as al -
antitrypsin
(ai-AT), secretory leukocyte protease inhibitor (SLPI) and pre-elafin leads to
the various
inflammatory disorders of COPD. Individuals that have a genetic deficiency of
the protease
inhibitor al-antitrypsin develop emphysema that increases in severity over
time (Laurrell,
C.B.; Erikkson, S Scand. I Clin. Invest. 1963 15, 132-140). An excess of HNE
is therefore
destructive, leading to the breakdown of pulmonary morphology with loss of
elasticity and
destruction of alveolar attachments of airways in the lung (emphysema) whilst
simultaneously increasing microvascular permeability and mucus hypersecretion
(chronic
bronchitis).
Several human neutrophil inhibitors have need disclosed so far in the art. In
particular, W02011/110858, W02011/110859 and WO 2014/009425 describe some
pyrimidine derivatives having human neutrophil elastase inhibitory properties
and their use
in therapy.
Although several FINE inhibitors have been disclosed so far as above reported,
there
is still a need for further HNE inhibitors. Particularly, there is still a
need for further HNE
inhibitors endowed with a high potency for FINE enzyme inhibition. The
identification of
further HNE inhibitors endowed with a high potency and which could be
advantageously
developed as an inhalation or oral treatment would be particularly
advantageous.
The present invention addresses the above mentioned need by providing the
compounds of the invention.

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
3
Brief description of the invention
This invention provides novel inhibitors of FINE, useful in the treatment of
diseases
or conditions in which FINE activity plays a role.
Detailed description of the invention
The invention there provides a compound of formula (I)
NC
)=B
Ris
0 N Me
XR3
(I)
wherein
A is -CH or ¨N-;
B is -CH or ¨N-;
D is -CH or ¨N-;
X is -CH or ¨N-;
R.3 is -CF3, CHF2 or halogen;
W is a 5 or 6-membered heteroaryl ring, optionally substituted with one or
more
substituents selected from -CN, -NO2, -0(C1-C6)alkyl, -(C1-C3)alky1-0-(Cl-
C6)alkyl, 4Ci-
C3)alkylamino, -(C3-C8)cycloalkyl, -OH, (C1-C3)alkyl, halo, -CF3 and ¨S02(Ci-
C4)alkyl;
Ri is H or is selected from the group consisting of -C(0)R4, -(CH2)t-(CHR6)n-
12.4, -(CH2)4CHR6)6S(02)-NR4R5, -S(02)-R4, -
(CH2)t-(CHR6)n-C(0)-
NR4R5, -(CH2)4CHR6)n-C(0)0-R4,
Ra is H or is selected from the group consisting of -(Ci-C6)alkyl, -(C2-
C6)alkyl-
NR7R8, -(C2-C6)alkyl-N R7R8R9, -(C2-C6)alky1-0-(Cl-C4)alkyl, -(C2-C6)alkyl-
S(02)(Ci-
C4)alkyl, ¨(CH2)t-aryl, ¨(CH2)t-(C3-C8)cycloalkyl, ¨(CH2)(C3-
C8)heterocycloalkyl and ¨

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
4
(CH2)t-heteroaryl, optionally substituted with one or more substituents
selected from -CN,
-NO2, -0 (C1-C6)alkyl, -(C1-C3)alky1-0-(C1-C6)alkyl, -(C1-C3)alkylamino, -(C3-
C8)cyclo alkyl, -OH, (C1-C3)alkyl, halo, -CF3 and ¨S 02(C 1 -C4)alkyl ;
n is 0 or an integer selected from 1, 2 or 3;
t is 0 or an integer selected from 1, 2 or 3;
R5 is H or -(C1-C6)alkyl;
R6 is H, -(C1-C6)alkyl, -CF3 or -CHF2;
R7 is H or -(C1-C6)alkyl;
R8 is H or -(C1-C6)alkyl;
R9 is H or -(C1-C6)alkyl;
wherein alternatively R4 and R5 together with the nitrogen atom they are
linked to,
may form a (C3-C8)heterocycloalkyl or heteroaryl ring system, optionally
substituted with
one or more substituents selected from -CN, -NO2, -0(C1-C6)alkyl, -(C1-
C3)alkyl-0-(Ci-
C6)alkyl, -(C1-C3)alkylamino, -(C3-C8)cycloalkyl, -OH, (C1-C3)alkyl, halo, -
CF3
and -S02(C1-C4)alkyl, wherein the heteroatoms of the (C3-C8)heterocycloalkyl
or
heteroaryl ring system are maximum three and selected from -N, -NH, -0, -S-
and -SO2;
R2 is H or is selected from the group consisting of, -SMe, ¨S(0)R7, ¨S(02)R7
and
halogen;
and pharmaceutically acceptable salts thereof.
The compounds of the invention may be used in the treatment or prevention of
diseases in which FINE is implicated, for example COPD, bronchiectasis,
chronic
bronchitis, lung fibrosis, pneumonia, ARDS, pulmonary emphysema, smoking-
induced
emphysema and CF.
Other aspects of the invention are (i) a pharmaceutical composition comprising
a
compound of the invention and one or more pharmaceutically acceptable carriers
or
excipients; and (ii) the use of a compound of the invention for the
manufacture of a
medicament for the treatment or prevention of a disease or condition in which
FINE is
implicated.

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
Unless otherwise specified, the term "substituted" as applied to any moiety
means
substituted with one or more substituents selected from -CN, -NO2, -0(Ci-
C6)alkyl, -(Ci-
C3)alky1-0-(Ci-C6)alkyl, -(C1-C3)alkylamino, -(C3-C8)cycloalkyl, -OH, (C1-
C3)alkyl, halo
(including fluoro, bromo and chloro), -CF3 and ¨S02(CI-C4)alkyl.
5 The term "halogen" includes fluorine, chlorine, bromine and iodine
atoms.
The term "(Cx-Cy)alkyl" wherein x and y are integers, refers to a straight or
branched
chain alkyl radical having x to y carbon atoms. Thus, when x is 1 and y is 6,
for example,
the term includes methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-
butyl, t-butyl, n-
pentyl and n-hexyl.
The term "(Cx-Cy)cycloalkyl", wherein x and y are integers, refers to
saturated
monocyclic, bicyclic or tricyclic hydrocarbon groups containing x to y ring
carbon atoms,
as appropriate, and wherein a carbon atom can be replaced by a group CO.
Examples
include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl,
adamantyl.
The term "heterocycloalkyl" relates to a saturated mono-, bi- or
tri-cyclic non-aromatic radical containing one or more heteroatoms selected
from S, SO,
SO2, N, NH and 0 and wherein a carbon atom may be replaced by a group CO. The
definition bicyclic heterocyclic systems, refers tofused, spiro and bridged
bicyclic systems,
such as for example a quinuclidine ring. In particular, the term "(Cx-
Cy)heterocycloalkyl"
refers to monocyclic (Cx-Cy)cycloalkyl groups, in which at least one ring
carbon atom is
replaced by a heteroatom (e.g. N, NH, S, SO, SO2 or 0). Examples of (Cx-
Cy)heterocycloalkyl include pyrrolidinyl, thiazolidinyl, piperazinyl,
piperidinyl,
morpholinyl, thiomorpholinyl.
The term "aryl" refers to a mono- or bi-cyclic carbocyclic aromatic radical,
and
includes radicals having two monocyclic carbocyclic aromatic rings which are
directly
linked by a covalent bond. Illustrative of such radicals are phenyl, biphenyl
and naphthyl.
The term "heteroaryl" refers to a mono- or bi-cyclic aromatic radical
containing one
or more heteroatoms selected from S, N and 0, and includes radicals having two
such
monocyclic rings, or one such monocyclic ring and one monocyclic aryl ring,
which are

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
6
fused through a common bond. Illustrative examples of 5,6-membered heteroaryl
are
thienyl, fury!, pyrrolyl, imidazolyl, thiazolyl, isothiazolyl, pyrazolyl,
oxazolyl, isoxazolyl,
isothiazolyl, triazolyl, thiadiazolyl, oxadiazolyl, pyridinyl, pyridazinyl,
pyrimidinyl,
pyrazinyl, and triazinyl. Illustrative examples of 8-10-membered heteroaryl
are
benzothienyl, benzofuryl, benzimidazolyl, benzothiazolyl, benzisothiazolyl,
benzoxazolyl,
benzisoxazolyl, benzotriazolyl, indolyl and indazolyl.
The term "salt" includes base addition and acid addition salts.
The term "pharmaceutically acceptable salts" refers to derivatives of
compounds of
formula (I) wherein the parent compound is suitably modified by converting any
of the free
acid or basic group, if present, into the corresponding addition salt with any
base or acid
conventionally intended as being pharmaceutically acceptable.
Compounds of the invention which are acidic can form salts, including
pharmaceutically acceptable salts, with bases such as alkali metal hydroxides,
e.g. sodium
and potassium hydroxides; alkaline earth metal hydroxides e.g. calcium, barium
and
magnesium hydroxides; with organic bases e.g. N-methyl-D-glucamine, choline
tris(hydroxymethyl)amino-methane, L-arginine, L-lysine, N-ethyl piperidine,
dibenzylamine and the like. Basic compounds can form pharmaceutically
acceptable salts
with inorganic acids, e.g. with hydrohalogen acids such as hydrochloric or
hydrobromic
acids, sulphuric acid, nitric acid or phosphoric acid and the like, and with
organic acids e.g
acetic, tartaric, succinic, fumaric, maleic, malic, salicylic, citric,
methanesulfonic, p-
toluenesulfonic, benzoic, benzenesulfonic, glutamic, lactic, and mandelic
acids and the like.
Compounds which have quaternary nitrogen can also form quaternary salts with a

pharmaceutically acceptable counter-ion such as chloride, bromide, acetate,
formate, p-
toluenesulfonate, succinate, hemi-succinate, naphthalene bis-sulfonate,
methanesulfonate,
xinafoate, and the like.
Where the compounds of the invention have at least one stereogenic center,
they may
exist as enantiomers. When the compounds according to the invention possess
two or more
stereogenic centers, they may additionally exist as diastereoisomers. It is to
be understood

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
7
that all such isomers and mixtures thereof in any proportion are encompassed
within the
scope of the invention.
Accordingly, a compound of the present invention can be in the form of one of
the
possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, for
example, as
substantially pure geometric (cis or trans) isomers, diastereoisomers, optical
isomers
(antipodes), racemates or mixtures thereof
Any formula given herein is also intended to represent unlabeled forms as well
as
isotopically labeled forms of the compounds. Isotopically labeled compounds
have
structures depicted by the formulas given herein except that one or more atoms
are replaced
by an atom having a selected atomic mass or mass number. Substitution with
heavier
isotopes, particularly deuterium (i.e., 2H or D) may afford certain
therapeutic advantages
resulting from greater metabolic stability, for example increased in vivo half-
life or reduced
dosage requirements or an improvement in therapeutic index. It is understood
that
deuterium in this context is regarded as a sub stituent of a compound as
defined in the first
aspect. The concentration of such a heavier isotope, specifically deuterium,
may be defined
by the isotopic enrichment factor. The term "isotopic enrichment factor" means
the ratio
between the isotopic abundance and the natural abundance of a specified
isotope. If a
substituent in a compound of this invention is denoted deuterium, such a
compound has an
isotopic enrichment factor for each designated deuterium atom of at least 3500
(52.5%
deuterium incorporation at each designated deuterium atom), at least 4000 (60%
deuterium
incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000
(75%
deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at
least 6000
(90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation),
at least
6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium
incorporation), or at
least 6633.3 (99.5% deuterium incorporation).
Isotopically-labeled compounds as defined in the first aspect can generally be

prepared by conventional techniques known to those skilled in the art or by
processes
analogous to those described using appropriate isotopically-labeled reagents
in place of the

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
8
non-labeled reagents previously employed.
It is to be understood that all preferred groups or embodiments described here
below
for compounds of formula (I) can be combined among each other and apply
mutatis
mutandis.
In one embodiment, for compounds of formula (I) A, B, D and X are ¨CH.
In one embodiment, W is a 5-membered heteroaryl ring.
In another preferred embodiment, W is selected from the group consisting of
pyrazolyl, imidazolyl and thiazolyl.
In one preferred embodiment, R3 is -CF3.
In one preferred embodiment, R2 is H.
In one preferred embodiment, R2 is ¨S(02)R7 wherein R7 is H or -(C1-C6)alkyl.
In one preferred embodiment, R2 is halogen.
In another preferred embodiment, Ri is H.
In another preferred embodiment, 114 is selected from the list consisting of -
C(0)R4;
-(CH2)t-(CHR6)n-R4; -(CH2)t-(CHR6)nS(02)-NR4R5; -S(02)-R4; -(CH2)t-(CHR6)n-
C(0)-
NR4R5 and -(CH2)t-(CHR6)n-C(0)0-R4.
In another preferred embodiment, RI is -(CH2)t-(CHR6).-R4 wherein R4 is H or
is
selected in the group consisting of -(C1-C6)alkyl, -(C2-C6)alkyl-NR7R8 and -
(C2-C6)alkyl-
S(02)(C1-C4)alkyl; wherein R7 is H or -(C1-C6)alkyl; R8 is H or -(C1-C6)alkyl;
t is 0 or an
integer selected from 1, 2 or 3; n is 0 or an integer selected from 1, 2 or 3.
In another preferred embodiment, Ri is -(CH2)t-(CHR6),S(02)-NR4R5 wherein R4
is
H or is selected in the group consisting of -(C1-C6)alkyl and -(C3-
C8)cycloalkyl; Rs is H or
-(C1-C6)alkyl; t is 0 or an integer selected from 1, 2 or 3; n is 0 or an
integer selected from
1, 2 or 3.
In another preferred embodiment, Ri is -(CH2)t-(CHR6)n-C(0)0-R4wherein R4 is H
or -(C1-C6)alkyl, optionally substituted with one or more substituents
selected
from -CN, -NO2, -0(C1-C6)alkyl, -OH, (C1-C3)alkyl, halo and -CF3; R6 is H, -
(C1-C6)alkyl,
-CF3 or -CHF2; t is 0 or an integer selected from 1, 2 or 3; n is 0 or an
integer selected from

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
9
1,2 or 3.
In another preferred embodiment, Ri is -C(0)R4, wherein R4 is H or -(C1-
C6)alkyl,
optionally substituted with one or more substituents selected from CN, -NO2, -
0(C1-
C6)alkyl, -OH, halo and -CF3.
In another preferred embodiment, Ri is -(CH2)t-(CHR6).-C(0)-NR4R5 wherein R4
is
H or is selected in the group consisting of -(C1-C6)alkyl, optionally
substituted with one or
more groups selected from -0(C1-C6)alkyl, -(C1-C3)alkylamino, ¨S02(C1-C4)alkyl

and -OH or R4 is selected in the group consisting of ¨(CH2)t-(C3-
C8)cycloalkyl, ¨(CH2)t-
(C3-C8)heterocycloalkyl, -(C2-C6)alkyl-NR7R8, -(CH2)t-aryl, -(CH2)t-heteroaryl
and -(C2-
C6)alky1-N R7R8R9, optionally substituted with one or more groups selected
from -(Ci-
C6)alkyl, ¨S02(C1-C4)alkyl and -OH; 115 is H or -(Ci-C6)alkyl; or wherein
alternatively R4
and R5 together with the nitrogen atom they are linked to, may form a (C3-
C8)heterocycloalkyl or heteroaryl ring system, optionally substituted with one
or more
substituents selected from ¨OH and (C1-C3)alkyl, wherein the heteroatoms of
the (C3-
C8)heterocycloalkyl or heteroaryl ring system are maximum three and selected
from -N, -
NH, -0, -S- and -SO2; R6 is H, -(C1-C6)alkyl, -CF3 or -CHF2; R7 is H or -(C1-
C6)alkyl; R8
is H or -(C1-C6)alkyl; t is 0 or an integer selected from 1, 2 or 3; n is 0 or
an integer selected
from 1,2 or 3.
In another preferred embodiment, Ri is -S(02)-R4 wherein R4 is H or is
selected in
the group consisting of ¨(CH2)t-(C3-C8)cycloalkyl and -(Cl-C6)alkyl; t is 0 or
an integer
selected from 1, 2 or 3.
In one embodiment, a compound formula I is selected in the group consisting of

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
Ex. Chemical name
1 445- 5-Methyl-2-oxo- 1 43 -(trifluoromethyl)phenyl] -2,3 -dihydro- 1 H-
imidazol-
4-yll -1 H-pyrazol- 1 -yl)benzonitrile
2 445 -15 -Methyl-3 -[3 -(methylsulfonyl)propy1]-2-oxo-1-[3 -
(trifluoromethyl)-
phenyl] -2,3 -dihydro- 1 H-imidazol-4-yll - 1H-pyrazol- 1 -yl)benzonitrile
3 4-(5- {3 ,5-Dimethy1-2-oxo- 1 43-(trifluoromethyl)phenyl] -2,3 -dihydro-
1 H-
imidazol-4 -y1 -1 H-pyrazol-1 -yl)benzonitrile
4 Ethyl-5- [1 -(4-cyanopheny1)- 1 H-pyrazol-5-y1]-4-methyl-2-oxo-3 -[3-
(trifluoromethyl)phenyl] -2,3 -dihydro- 1 H-imidazole- 1 -carboxylate
5 4-(5 - {3 -Acetyl-5 -methyl-2-oxo-1 -(trifluoromethyl)phenyl]-2,3 -
dihydro- 1 H-
imidazol-4-yll -1 H-pyrazol- 1 -yl)benzonitrile
6 5- [1 -(4-Cyanopheny1)- 1 H-pyrazol-5-yl] -N-ethyl-4-methyl-2-oxo-3 -
[3 -
(trifluoromethyl)pheny1]-2,3 -dihydro-1 H-imidazol e-1 -carboxamide
7 5-[1 -(4-Cyanopheny1)- 1 H-pyrazol-5-yl]-N-(2-hydroxyethyl)-4-methyl-2-
oxo-3 -
[3 -(trifluoromethyl)phenyl]-2,3 -dihydro- 1 H-imidazo le- 1 -carboxamide
8 5-[1 -(4-Cyanopheny1)-1 H-pyrazol-5 -y1]-N-(3 -hydroxypropy1)-4-methy1-
2 -oxo-
3 - [3 -(trifluoromethyl)phenyl]-2,3 -dihydro- 1H-imidazole- 1 -carboxamide
9 5- [1 -(4-Cyanopheny1)-1H-pyrazol-5-yl] -N- -(dimethylamino)propyl] -4-
methy1-2 -oxo-3 -(trifluoromethyl)phenyl] -2,3 -dihydro-1H-imidazole-l-
carboxamide
10 4-(5- {3-Butanoy1-5-methyl-2-oxo- 1 -[3 -(trifluoromethyl)phenyl]-2,3 -
dihydro-
1H-imidazol-4-y1 -1 H-pyrazol-1 -yl)benzonitrile
11 5- [1 -(4-Cyanopheny1)-1H-pyrazol-5-yl] -N-(2-methoxyethyl)-4-methyl-2-
oxo-3 -
[3 -(trifluoromethyl)phenyl]-2,3 -dihydro- 1H-imidazole- 1 -carboxamide
12 5- [1 -(4-Cyanopheny1)- 1 H-pyrazol-5-yl] -N- [2-(dimethylamino)ethy1]-
4-methy1-
2-oxo-3 -(trifluoromethyl)phenyl] -2,3 -dihydro- 1 H-imidazo le-1 -carboxamide
13 5-[1 -(4-Cyanopheny1)- 1 H-pyrazol-5-y1]-4-methyl-N- [2-
(methylsulfonypethyl] -
2-oxo-3- [3 -(trifluoromethyl)phenyl] -2,3 -dihydro- 1 H-imidazole- 1 -
carboxamide
14 5-[1 -(4-Cyanopheny1)- 1 H-pyrazol-5-y1]-4-methyl-2-oxo-N-
(tetrahydrofuran-3 -
y1)-3 43 -(trifluoromethyl)phenyl] -2,3 -dihydro- 1 H-imidazole- 1 -
carboxamide
5-[l -(4-Cyanopheny1)-1H-pyrazol-5-y1]-4-methy1-2-oxo-N-[(3 S)-
tetrahydrfuran-3 -y1]-3 -[3 -(trifluoromethyl)phenyl]-2,3 -dihydro- 1H-
imidazole-
1 -carboxamide
(continued)

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
11
16 5-[1-(4-Cyanopheny1)-1H-pyrazol-5-y1]-4-methy1-2-oxo-N- [(3R)-
tetrahydrofuran-3-yl] -3- [3-(trifluoromethyl)pheny1]-2,3-dihydro-1H-imidazole-

1-carboxamide
17 5- [1-(4-Cyanopheny1)-1H-pyrazol-5-y1]-4-methyl-N4(1-methyl-1H-pyrazol-
3-
yOmethyl] -2-oxo-343-(trifluoromethyl)phenyl] -2,3-dihydro-1H-imidazole-1-
carboxamide
18 Methyl-5- [1-(4-cyanopheny1)-1H-pyrazol-5-yl] -4-methyl-2-oxo-3- [3-
(trifluoromethyl)phenyl] -2,3-dihydro-1H-imidazole-1-carboxylate
19 4-(5- {5-Methy1-2-oxo-3-pentanoy1-143-(trifluoromethyl)pheny11-2,3-
dihydro-
1H-imidazol-4-yll -1H-pyrazol-1-yl)benzonitrile
20 tert-Butyl- {5- [1-(4-cyanopheny1)-1H-pyrazol-5-yl] -4-methyl-2-oxo-3-
[3 -
(trifluoromethyl)phenyl] -2,3-dihydro-1H-imidazol-1-yll acetate
21 5-[1-(4-Cyanopheny1)-1H-pyrazol-5-y1]-N-ethyl-N,4-dimethyl-2-oxo-3- [3-
(trifluoromethyl)pheny1]-2,3-dihydro-111-imidazole-1-carboxamide
22 {541-(4-Cyanopheny1)-1H-pyrazol-5-yl] -4-methyl-2-oxo-3- [3-
(trifluoromethyl)phenyl] -2,3-dihydro-1H-imidazol-1-yll acetic acid
23 2- {511-(4-Cyanopheny1)-1H-pyrazol-5-y1]-4-methy1-2-oxo-343-
(trifluoromethyl)phenyl]-2,3-dihydro-1H-imidazol-1-yll -N-methylacetamide
24 Benzenesulfonate 3 -[( {5- [1-(4-cyanopheny1)-1H-pyrazol-5-y1]-4-methy1-
2-oxo-
3- [3-(trifluoromethyl)pheny1]-2,3-dihydro-1H-imidazol-1-ylIcarbonyl)amino] -
N,N,N-trimethylpropan-1-aminium
25 Benzenesulfonate 24( {5- [1-(4-cyanopheny1)-1H-pyrazol-5-yl] -4-methy1-
2-oxo-
3- [3 -(trifluoromethyl)phenyl] -2,3-dihydro-1H-imidazol-1-ylIcarbonypamino] -

N,N,N-trimethylethanaminium
26 4-(5- {3-Buty1-5-methy1-2-oxo-1-[3-(trifluoromethypphenyl]-2,3-dihydro-
1H-
imidazol-4-yll -1H-pyrazol-1-yl)benzonitrile
27 4-(5- {5-Methy1-3-(methylsulfony1)-2-oxo-1- [3-(trifluoromethyl)phenyl]
-2,3-
dihydro-1H-imidazol-4-yll -1H-pyrazol-1-yl)benzonitrile
28 4- {545-Methy1-3-(1-methyl-buty1)-2-oxo-1-(3-trifluoromethyl-pheny1)-
2,3-
dihydro-1H-imidazol-4-ylipyrazol-1-yll -benzonitrile
29 4- {5- [3-(2-Dimethylamino-ethyl)-5-methyl-2-oxo-1-(3-trifluoromethyl-
pheny1)-2,3-dihydro-1H-imidazol-4-y11-pyrazol-1-yll -benzonitrile
(continued)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
12
30 4- {5-[3-(3-Dimethylamino-propy1)-5-methy1-2-oxo-1-(3-trifluoromethyl-
phenyl)-2,3-dihydro-lH-imidazol-4-y1]-pyrazol-1-y1} -benzonitrile
31 4- {5- [5-Methy1-2-oxo-3-(propane-l-sulfony1)-1-(3-trifluoromethyl-
pheny1)-2,3-
dihydro-1H-imidazol-4-y1]-pyrazol-1-yll -benzonitrile
32 4- {5- [3 -Cyclopropanesulfony1-5-methy1-2-oxo-1-(3-trifluoromethyl-
pheny1)-
2,3-dihydro-1H-imidazol-4-yl]-pyrazol-1-yll -benzonitrile
33 5- [2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-
trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-sulfonic acid ethylamide
34 5- [2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-
trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-sulfonic acid cyclopentylamide
35 5- [2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-
trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-sulfonic acid cyclopropylamide
36 4- {5- [5-Methy1-2-oxo-3-(2-oxo-2-pyrrolidin-1-yl-ethyl)-1-(3-
trifluoromethyl-
pheny1)-2,3-dihydro-1H-imidazol-4-y1]-pyrazol-1-yll -benzonitrile
37 2- [5- [2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-
trifluoromethyl-pheny1)-2,3-dihydro-imidazol-1-yll -N,N-dimethyl-acetamide
38 4- {5- [5-Methy1-3- [2-(4-methyl-piperazin-1-y1)-2-oxo-ethyl]-2-oxo-1-(3-

trifluoromethyl-pheny1)-2,3-dihydro-11-1-imidazol-4-y1}-pyrazol-1-yll -
benzonitrile
39 2- [542-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-
trifluoromethyl-pheny1)-2,3-dihydro-imidazol-1-y1]-N-(3-dimethylamino-
propy1)-acetamide formic acid salt
40 2- [542-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-
trifluoromethyl-pheny1)-2,3-dihydro-imidazol-1-y1]-1\1-cyclobutyl-N-methyl-
acetamide
41 5- [2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-
trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid cyclobutylamide
42 5- [2-(4-Cyano-pheny1)-2H-pyrazol-3-y11-4-methy1-2-oxo-3-(3-
trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid cyclopropylamide
(continued)

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
13
43 542-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid (1-ethanesulfonyl-piperidin-4-
y1)-amide
44 5-[2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid (tetrahydro-pyran-4-y1)-amide
45 542-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid cyclopentylamide
46 5-[2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid ((R)-1-methyl-pyrrolidin-3-
ye-amide
47 5-[2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid ((S)-1-methyl-pyrrolidin-3-
y1)-amide
48 Mixture of 542-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methyl-2-oxo-3-(3-
trifluoromethyl-pheny1)-2,3-dihydro-imidazole-1-carboxylic acid ((3R,4S)-4-
hydroxy-tetrahydro-furan-3-y1)-amide and 542-(4-Cyano-pheny1)-2H-pyrazol-
3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-pheny1)-2,3-dihydro-imidazole-1-
carboxylic acid ((3S,4R)-4-hydroxy-tetrahydro-furan-3-y1)-amide
49 5-[2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methyl-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid (pyridin-2-ylmethyp-amide
50 5-[2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid (pyridin-3-ylmethyp-amide
51 5-[2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid (pyridin-4-ylmethyl)-amide
52 5-[2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methyl-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid (pyrazin-2-ylmethyl)-amide
53 542-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid ((1R,2R)-
2-hydroxy-
cyclopenty1)-amide
54 5-[2-(4-Cyano-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-trifluoromethyl-
pheny1)-2,3-dihydro-imidazole-1-carboxylic acid (1-
methyl-piperidin-4-
ylmethyp-amide
(continued)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
14
55 3 -Methanesulfony1-4- {5- [5-methy1-2-oxo-1-(3-trifluoromethyl-pheny1)-
2,3-
dihydro-1H-imidazol-4-yl] -pyrazol-1-y1) -benzonitrile
56 5- [2-(2-Bromo-4-cyano-phenyl)-2H-pyrazol-3 -yl] -4-methyl-2-oxo-3 -(3 -

trifluoromethyl-pheny1)-2,3 -dihydro-imidazole-l-carboxylic acid
cyclopentylamide
57 5- [1-(2-Bromo-4-cyano-pheny1)-1H-pyrazol-3 -y1]-4-methyl-2-oxo-3 -(3-
trifluoromethyl-pheny1)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide
58 5- [2-(4-Cyano-2-fluoro-pheny1)-2H-pyrazol-3-y1]-4-methy1-2-oxo-3-(3-
trifluoromethyl-pheny1)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide
59 445'-Methy1-2'-oxo-1'-(3-trifluoromethyl-pheny1)-2',3'-dihydro-1'H-
[2,41biimidazoly1-1-y1]-benzonitrile
60 1-(4-Cyano-pheny1)-5'-methy1-2'-oxo-1'-(3-trifluoromethyl-pheny1)-1',2'-
dihydro-1H42,41biimidazoly1-3'-carboxylic acid cyclopentylamide
61 1-(4-Cyano-pheny1)-51-methy1-2'-oxo-1'-(3-trifluoromethyl-pheny1)-1',2'-
dihydro-111- [2,41 biimidazoly1-3'-carboxylic acid cyclobutylamide
62 1-(4-Cyano-pheny1)-5'-methy1-2'-oxo-1'-(3-trifluoromethyl-pheny1)-1',2'-
dihydro-1H42,41biimidazoly1-3'-carboxylic acid 4-methanesulfonyl-
benzylamide
63 5- [2-(4-Cyano-phenyl)-2H-[1,2,4]triazol-3 -yl] -4-methyl-2-oxo-3 -(3 -
trifluoromethyl-pheny1)-2,3 -dihydro-imidazole-l-carboxylic acid
cyclopentylamide
and pharmaceutically acceptable salts thereof.
The present invention is also directed to a process for the preparation of a
compound
of formula (I)
NC
)=B
R W
(:)N Me
R,
(I)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
by reacting a compound of formula (IV)
NCO
)(R3
(IV)
with prop-2-ynylamine to obtain a compound of formula (V)
5
HN
o
NH

XR3
(V)
The invention is also directed to a process for the preparation of a compound
of
formula (I) by reacting a compound of formula (V) following treatment with a
base to
obtain a compound of formula (VI)
O')Me
XR3
10 (VI)
The invention is also directed to a process for the preparation of a compound
of
formula (I) by reacting a compound of formula (VI) with an alkylating agent to
obtain a
compound of formula (VII)
N
0 N Me
(VII)
15 The invention is also directed to a process for the preparation of a
compound of

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
16
formula (I) by reacting a compound of formula (VII) with a brominating agent
to obtain a
compound of formula (VIII)
Br
N
0 N Me
(VIII)
The invention is also directed to a process for the preparation of a compound
of
formula (I) by reacting a compound of formula (VIII) with a compound of
formula (IX)
R2
,N
N


(nBu)3Sn
(IX)
to obtain a compound of formula (II)
NC--e-572
,
N
N
0 N Me
XR3
(II)
The invention also provides a pharmaceutical composition comprising a compound

of formula (I) together with one or more pharmaceutically acceptable carriers
or excipients.
The invention also provides a pharmaceutical composition comprising a compound

of formula (I) which is adapted for oral administration or administration by
the pulmonary
route.
The invention also provides a compound of formula (I) for use in the treatment
of,

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
17
or for use in the manufacture of a medicament for the treatment of a disease
or condition in
which HNE is implicated.
The invention also provides a method of treatment of a disease or condition in
which
HNE is implicated, comprising administering to a subject suffering from such
disease an
effective amount of a compound of formula (I).
The invention also provides a compound of formula (I) for use, or a method of
treatment, wherein the disease or condition is COPD, bronchiectasis, chronic
bronchitis,
lung fibrosis, pneumonia, ARDS, pulmonary emphysema, smoking-induced emphysema

or cystic fibrosis.
The invention also provides a compound of formula (I) for use, or a method of
treatment, wherein the disease or condition is asthma, rhinitis, psoriasis,
atopic dermatitis,
non-atopic dermatitis, Crohn's disease, ulcerative colitis, or irritable bowel
disease.
The therapeutic utility of the present compounds is pertinent to any disease
that is
known to be at least partially mediated by the action of human neutrophil
elastase. For
example, the present compounds may be beneficial in the treatment of COPD, CF,
bronchiectasis, ARDS, pulmonary emphysema, pneumonia and lung fibrosis.
Compounds of the invention are useful for treatment of inflammatory
respiratory
disorders, for example asthma (mild, moderate or severe), steroid resistant
asthma,
bronchitis, COPD, CF, pulmonary edema, pulmonary embolism, pneumonia,
pulmonary
sarcoidosis, pulmonary emphysema, silicosis, pulmonary fibrosis, pulmonary
hypertension, respiratory failure, ARDS, emphysema, chronic bronchitis,
tuberculosis,
aspergillosis and other fungal infections, hypersensitivity pneumonitis,
vasculitic and
thrombotic disorders of the lung vasculature, antitussive activity including
treatment of
chronic cough associated with inflammatory and secretory conditions of the
airways,
infection due to respiratory syncytial virus, influenza, coronavirus
(including severe acute
respiratory syndrome, SARS) and adenovirus, bronchiectasis and lung cancer.
The invention also concerns pharmaceutical formulations comprising, as an
active
ingredient, a compound of the invention. Other compounds may be combined with

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
18
compounds of this invention for the prevention and treatment of inflammatory
diseases of
the lung. Thus the invention also concerns pharmaceutical compositions for
preventing and
treating inflammatory diseases of the lung comprising a therapeutically
effective amount
of a compound of the invention and one or more other therapeutic agents.
Suitable therapeutic agents for a combination therapy include: (1) a cortico
steroid,
for example budesonide, beclomethasone, beclomethasone (e.g., as the mono or
the
dipropionate ester), flunisolide, fluticasone (e.g. as the propionate or
furoate ester),
Ciclesonide, mometasone (e.g. as the furoate ester), mometasone desonide,
rofleponide,
hydrocortisone, prednisone, prednisolone, methyl prednisolone, naflocort,
deflazacort,
halopredone acetate, fluocinolone acetonide, fluocinonide, clocortolone,
tipredane,
prednicarbate, alclometasone dipropionate, halometasone, rimexolone, deprodone

propionate, triamcinolone, betamethasone, fludrocoritisone,
desoxycorticosterone,
rofleponide, etiprednol dicloacetate and the like. Steroid drugs can
additionally include
steroids in clinical or pre-clinical development for respiratory diseases such
as GW-685698,
GW-799943, GSK 870086, QAE397, NCX-1010, NCX-1020, NO-dexamethasone, PL-
2146, NS-126 (formerly ST-126). Steroid drugs can also additionally include
next
generation molecules in development with reduced side effect profiles such as
selective
glucocorticoid receptor agonists (SEGRAs), including ZK-216348 and AZD5423;
(2) a132-
adrenoreceptor agonist, such as albuterol, bambuterol, terbutaline, fenoterol,
formoterol,
formoterol fumarate, salmeterol, salmeterol xinafoate, arformoterol,
arfomoterol tartrate,
indacaterol (QAB-149), carmoterol, BI 1744 CL, GSK159797 (milveterol),
GSK59790,
GSK159802, GSK642444 (vilanterol), GSK678007, GSK96108, clenbuterol,
procaterol,
bitolterol, LAS100977 (abediterol), BI1744CL (olodaterol) and brodxaterol; (3)
a
leukotriene modulator, for example montelukast, zafirlukast or pranlukast; (4)
anticholinergic agents, for example selective muscarinic-3 (M3) receptor
antagonists such
as ipratropium bromide, tiotropium, tiotropium bromide (Spiriva ),
glycopyrronium
bromide, aclidinium bromide, LAS34273, GSK656398, GSK233705, GSK 573719
(umeclidinium), LAS35201, QAT370 and oxytropium bromide; (5) phosphodiesterase-
IV

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
19
(PDE-IV) inhibitors, for example roflumilast, cilomilast or theophylline; (6)
an antitussive
agent, such as codeine or dextramorphan; and (7) a non-steroidal anti-
inflammatory agent
(NSAID), for example ibuprofen or ketoprofen; (8) a mucolytic, for example N
acetyl
cysteine or fudostein; (9) a expectorant/mucokinetic modulator, for example
ambroxol,
hypertonic solutions (e.g. saline or mannitol) or surfactant; (10) a peptide
mucolytic, for
example recombinant human deoxyribonoclease I (dornase-alfa and rhDNase) or
helicidin;
(11) antibiotics, for example azithromycin, tobramycin and aztreonam; and (12)
p38
Mitogen Activated Protein (MAP) kinase inhibitors, such as GSK 856553 and GSK
681323; (12) inhibitors of Janus Kinases (JAK) such as CP-690550 or GLPG0634;
(13)
Spleen Tyrosine Kinase (SYK) inhibitors such as R406, R343 or PRT062607; (14)
inhibitors of delta and/or gamma isoforms of Phosphatidylinositol 3-kinase
(PI3K).; (15)
anti-retroviral agents such as ribavirin, zanamivir or laninamivir; (16) PPAR-
y agonists
such as pioglitazone and rosiglitazone.
In one aspect, the invention provides for the use of inhaled administration of
compounds of the invention in combination with other anti-inflammatory drugs
and
bronchodilator drug combinations (i.e. triple combination product), including
but not
limited to salmeterol xinafoate/fluticasone propionate (Advair/Seretidet),
vilanterol/fluticasone furoate (BREO ELLIPTATm), formoterol
fumarate/budesonide
(Symbicortg), formoterol fumarate/mometasone furoate,
formoterol
fumarate/beclometasone dipropionate (Foster ), formoterol fumarate/fluticasone

propionate (FlutiForme), Indacaterol/mometasone furoate, Indacaterol/QAE-397,
GSK159797/GSK 685698, GSK159802/GSK 685698, GSK642444/GSK 685698,
formoterol fumarate/ciclesonide, arformoterol tartrate/ciclesonide.
In another aspect, the invention provides for the use of inhaled
administration of
compounds of the invention in combination with other bronchodilator drug
combinations,
particularly 132 agonist/M3 antagonist combinations (i.e. triple combination
product),
including but not limited to salmeterol xinafoate/tiotropium bromide,
formoterol
fumarate/tiotropium bromide, formoterol fumarate/ glycopyrrolate (PT003), BI
1744

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
CL/tiotropium bromide, indacaterol/NVA237, indacterol/QAT-370, formoterol/
LAS34273, umeclidinium/vilanterol (AnoroTm), GS
K159797/GSK 573719,
GSK159802/GSK 573719, GSK642444/GSK 573719, GSK159797/GSK 233705,
GSK159802/GSK 233705, GSK642444/GSK 233705.
5 The
weight ratio of the first and second active ingredients may be varied and will
depend upon the effective dose of each ingredient. Generally, an effective
dose of each will
be used.
The magnitude of prophylactic or therapeutic dose of a compound of the
invention
will, of course, vary with the nature of the severity of the condition to be
treated and with
10 the particular compound and its route of administration, and will
generally be determined
by clinical trial as required in the pharmaceutical art. It will also vary
according to the age,
weight and response of the individual patient. In general, the daily dose
range will lie within
the range of from about 0.001 mg to about 100 mg per kg body weight of a
mammal,
preferably 0.01 mg to about 50 mg per kg, and most preferably 0.1 to 10 mg per
kg, in
15 single or divided doses. On the other hand, it may be necessary to use
dosages outside these
limits in some cases.
Another aspect of the invention provides pharmaceutical compositions
comprising a
compound of the invention and a pharmaceutically acceptable carrier. The term
"composition", as in pharmaceutical composition, is intended to encompass a
product
20 comprising the active ingredient(s), and the inert ingredient(s)
(pharmaceutically
acceptable excipients) that make up the carrier, as well as any product which
results,
directly or indirectly, from combination, complexation or aggregation of any
two or more
of the ingredients, or from dissociation of one or more of the ingredients, or
from other
types of reactions or interactions of one or more of the ingredients.
Accordingly, the
pharmaceutical compositions of the invention encompass any composition made by
mixing
a compound of the invention, additional active ingredient(s), and
pharmaceutically
acceptable excipients.
The pharmaceutical compositions of the invention comprise a compound of the

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
21
invention as an active ingredient or a pharmaceutically acceptable salt
thereof, and may
also contain a pharmaceutically acceptable carrier and optionally other
therapeutic
ingredients. The term "pharmaceutically acceptable salts" refers to salts
prepared from
pharmaceutically acceptable non-toxic bases or acids including inorganic bases
or acids
and organic bases or acids.
Any suitable route of administration may be employed for providing a mammal,
especially a human, with an effective dosage of a compound of the invention.
In therapeutic
use, the active compound may be administered by any convenient, suitable or
effective
route. Suitable routes of administration are known, and include oral,
intravenous, rectal,
parenteral, topical, ocular, nasal, buccal and pulmonary (by inhalation).
Compositions suitable for administration by inhalation are known, and may
include
carriers and/or diluents that are known for use in such compositions. The
composition may
contain 0.01-99% by weight of active compound. Preferably, a unit dose
comprises the
active compound in an amount of 1 jig to 10 mg.
The most suitable dosage level may be determined by any known suitable method.
It will be understood, however, that the specific amount for any particular
patient will
depend upon a variety of factors, including the activity of the specific
compound that is
used, the age, body weight, diet, general health and sex of the patient, time
of
administration, the route of administration, the rate of excretion, the use of
any other drugs,
and the severity of the disease to be treated.
For delivery by inhalation, the active compound is preferably in the form of
microparticles. They may be prepared by a variety of techniques, including
spray-drying,
freeze-drying and micronisation.
By way of example, a composition of the invention may be prepared as a
suspension
for delivery from a nebuliser or as an aerosol in a liquid propellant, for
example for use in
a pressurised metered dose inhaler (PMDI). Propellants suitable for use in a
PMDI are
known to the skilled person, and include CFC-12, HFA-134a, HFA-227, HCFC-22
(CC12F2) and HFA-152 (CH4F2 and isobutane).

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
22
In a preferred embodiment, a composition of the invention is in dry powder
form,
for delivery using a dry powder inhaler (DPI). Many types of DPI are known.
Microparticles for delivery by administration may be formulated with
excipients that
aid delivery and release. For example, in a dry powder formulation,
microparticles may be
formulated with large carrier particles that aid flow from the DPI into the
lung. Suitable
carrier particles are known, and include lactose particles; they may have a
mass median
aerodynamic diameter of greater than 90 pm.
In the case of an aerosol-based formulation, a preferred composition is:
Compound of the invention 24 mg / canister
Lecithin, NF Liq. Conc. 1.2 mg / canister
Trichlorofluoromethane, NF 4.025 g / canister
Dichlorodifluoromethane, NF 12.15 g / canister.
The compounds of the invention may be used in combination with other drugs
that
are used in the treatment/prevention/suppression or amelioration of the
diseases or
conditions for which present compounds are useful. Such other drugs may be
administered,
by a route and in an amount commonly used therefore, contemporaneously or
sequentially
with a compound of the invention. When a compound of the invention is used
contemporaneously with one or more other drugs, a pharmaceutical composition
containing
such other drugs in addition to the compound of the invention is preferred.
Accordingly,
the pharmaceutical compositions of the invention include those that also
contain one or
more other active ingredients, in addition to a compound of the invention.
The agents of the invention may be administered in inhaled form. Aerosol
generation
can be carried out using, for example, pressure-driven jet atomizers or
ultrasonic atomizers,
preferably using propellant-driven metered aerosols or propellant-free
administration of
micronized active compounds from, for example, inhalation capsules or other
"dry powder"
delivery systems.
The active compounds may be dosed as described depending on the inhaler system

used. In addition to the active compounds, the administration forms may
additionally

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
23
contain excipients, such as, for example, propellants (e.g. Frigen in the case
of metered
aerosols), surface-active substances, emulsifiers, stabilizers, preservatives,
flavorings,
fillers (e.g. lactose in the case of powder inhalers) or, if appropriate,
further active
compounds.
For the purposes of inhalation, a large number of systems are available with
which
aerosols of optimum particle size can be generated and administered, using an
inhalation
technique which is appropriate for the patient. In addition to the use of
adaptors (spacers,
expanders) and pear-shaped containers (e.g. Nebulatort, Volumatic0), and
automatic
devices emitting a puffer spray (Autohaler0), for metered aerosols, in
particular in the case
of powder inhalers, a number of technical solutions are available (e.g.
Diskhalere,
Rotadiske, Turbohaler0 or the inhalers for example as described EP-A-0505321).

Procedure for the preparation of compounds of formula (I)
The invention further provides a process for the preparation of a compound of
formula (I) or a pharmaceutically acceptable salt or quaternary salt thereof
as defined above,
according to routes illustrated below in Schemes A and B.
The skilled person may introduce, where appropriate, suitable variations to
the
conditions specifically described in the experimental in order to adapt the
synthetic routes
to the provision of further compounds of the invention. Such variations may
include use of
appropriate starting materials to generate different compounds, changes in the
solvent and
temperature of reactions, replacements of reactives with analogous chemical
role,
introduction or removal of protection/deprotection stages of functional groups
sensitive to
reaction conditions and reagents.
Also, introduction or removal of specific synthetic steps oriented to further
functionalization of the chemical scaffold may be contemplated and is included
within the
scope of the present invention. Processes which can be used and are described
and reported
in Examples and Schemes, should not be viewed as limiting the scope of the
synthetic
methods available for the preparation of the compounds of the invention.
Compounds used as starting materials or intermediates may be commercially

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
24
available, their preparation may be specifically described in the literature
or they may be
prepared according to known methods. In some instances, procedures for the
preparation
of intermediates or starting materials may be also provided in the
experimental part.
The process described is particularly advantageous as it is susceptible of
being
properly modulated, through any known proper variant.
From all of the above, it should be clear that any of the described groups may
be
present as such or in any properly protected form.
In particular, functional groups present in the intermediate and compounds and

which could generate unwanted side reaction and by-products, need to be
properly
protected before the alkylation, acylation, coupling or sulfonylation takes
place. Likewise,
subsequent deprotection of those same protected groups may follow upon
completion of
the said reactions.
In the present invention, unless otherwise indicated, the term "protecting
group"
designates a protective group adapted to preserve the function of the group it
is bound to.
Typically, protective groups are used to preserve amino, hydroxyl, or carboxyl
functions.
Appropriate protecting groups may thus include, for example, benzyl,
benzyloxycarbonyl,
t-butoxycarbonyl, alkyl or benzyl esters or the like, which are well known
[see, for a general
reference, T.W. Green; Protective Groups in Organic Synthesis (Wiley, N.Y.
1981)].
Likewise, selective protection and deprotection of any of the said groups, for
instance including carbonyl, hydroxyl or amino groups, may be accomplished
according to
known methods
From all of the above, it should be clear that the above process,
comprehensive of
any variant thereof for the preparation of suitable compounds of the
invention, may be
conveniently modified so that to adapt the reaction conditions to the specific
needs, for
instance by choosing appropriate condensing agents, solvents and protective
groups, as the
case may be.
In the following Schemes, for compounds of formula (II) to (Xin) and (IB) to
(IF), unless otherwise indicated, groups A, B, D, X, W, R1 to R3 have the same
meanings

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
as described for compounds of formula (I) above.
In one aspect of the invention, a process for the preparation of compounds of
the
invention (TB), i.e. compounds of formula (I) wherein W = pyrazole (or as
defined above),
A, B, D, X and RI to R3 are as defined above, may be prepared according to
general
5 synthetic routes reported in Scheme A here below.
Scheme A
Method F
i) R1-Hal, MeCN
ii) Na0H(aq), MeCN
NCyB,R2
wherein R1=-(CH2)401-1R6)n-R4; A. ,k N
-(CH2)t(CHR6)n-CONR4R5; D 1)
and
for all R1 meanings, t and n R1 ---
N
can't be simultaneously 0 \ NCyBrõ,
R2
0 N Me A, ,..1 -N
o el _ Method A ) D 11_.)
----
X R3 NaH, DMF R40 N\
0-COCI 0 N Me
wherein R1=-(CH2)t(CHR6)n-R4; R4 ___ ._
-(CH2)t(CHR6)n-CONR4R5; (IC) (ID)
and
for all R1 meanings, t and n
'LI
can't be simultaneously 0 XR3
Method D
NaH, THF or DMF
R1-Hal NCyBR2
A, ,k N
Method E o DN f
N p
p R2 NaH, DMF
NC 1' NC.--
_.(13-fR2 , R4--g ---
\ N N R4-
S(02)CI Cr '
AD N. ) A--,[/---N \
1M HCI(aq) HN (IG)
_?..._ o N Me
Acetone \
¨0--NN Me ____________ ) 0 N Me ;---1
"XR3
)--- %L--
I I
)(R3 '-)( R3 NC .7.13
j(.,õ R2
Method B
1
(II) (IB) NaH, THF
R4COCI A,D 12)'I\
0
02N io 0 ). ¨
R4 N
cyjlci Et3N, DCM \
ON-Me
(1E)
¨ ))
NCyl3R2 _ X R3
02N 0 0o DN rµp
0 N Me NC.B.,.R2
). ----
A, -N
\ Method C o D _II i
R4R5NH R4\ ..1... "
N N (IF)
/ \
I ___________ , R5/ ON me
R3
(111) ei
X R3

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
26
Compounds of formula (IB) may be prepared from compounds of formula (II) by
reaction with an acid such as hydrochloric acid in a solvent such as acetone
at temperature
of from RT to 60 C. Compounds of formula (IB) may be converted into compounds
of
formula (IC) according to Method D, by reaction with an alkyl halide of
formula Ri-Hal
(Hal = Cl, Br, I) or Ri-X' wherein X' is an appropriate leaving group (e.g.
X'= tosylate) in
a solvent such as THF following addition of a base such as sodium hydride or
lithium
hexamethyldisilazide at a temperature of from 0 C to 50 C. Alternatively,
compounds of
formula (IC) can be prepared according to Method F, by direct alkylation with
Ri-Hal or
Ri-X' at a temperature of up to 120 C in a sealed vessel, followed by
hydrolysis of the
intermediate quaternary imidazolium compound with a base such as aqueous
sodium
hydroxide in a solvent such as acetonitrile.
Compounds of formula (ID), (IG) and (IE) can be prepared from compounds of
formula (IB) by addition of a base such as sodium hydride followed by reaction
with a
chloroformate of formula R4CO2C1, or R4S02C1 or acid chloride of formula
R4C0C1,
according to Methods A, E and B respectively, in a suitable solvent such as
THF or DMF
at temperatures of from 0 C to 60 C. Compounds of formula (IF) can be prepared
from
compounds of formula (IB) following the generation of the nitro-chloroformate
intermediate (III) and reaction with a suitable amine R4R5NH, in a solvent
such as DCM at
a temperature of from 0 C to 40 C.
Compounds of formula (II) can be prepared according to Scheme B below from
compounds of formula (IV):

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
27
Scheme B
NCO 1121\1, NaH Et30+BF4- OMe0 NHQMe
MeCN MeCN DCM
X R,X R,
(IV) (V) (VI) (VII)
B R2
Br
NBS Pd(PPh3)2C12
MeCN Dioxane A.DN
0 N Me
B R2
0 N Me
R3
(nBu),Sn
R3
(VIII) (IX) 00
Compounds of formula (V) may be obtained from compounds of formula (IV) by
reacting with prop-2-ynylamine in a suitable solvent such as DCM at a suitable
temperature
of between 0 C and RT. Compounds of formula (VI) can be prepared from
compounds of
formula (V) following treatment with a suitable base such as sodium hydride in
a suitable
solvent such as acetonitrile or THF (or mixtures thereof). The conversion of
compounds of
formula (VI) to compounds of formula (VII) can be effected by reaction with a
suitable
alkylating agent such as trimethoxonium tetrafluoroborate, in a suitable
solvent such as
DCM at a suitable temperature of between 0 C and RT. Compounds of formula
(VIII) can
be prepared from compounds of formula (VII) using a brominating agent such as
N-bromosuccinimide or bromine in a suitable solvent such as acetonitrile at a
suitable
temperature of between 0 C and RT. Compounds of formula (II) can be prepared
from
compounds of formula (VIII) using known C-C and C-N cross-coupling
methodologies
described in the literature using a suitably functionaliszed linking group "W"
such as aryl,
heteroaryl, cycloalkyl or heterocycloalkyl boronic acid/boronate ester,
stannane or zincate.
Typical reaction conditions consist of the use of an organometallic reactant
such as
4-(5-tributylstannanyl-pyrazol-1-y1)-benzonitrile (IX) and a palladium source
such as
bis(triphenylphosphine)palladium (II) dichloride and a solvent such as dioxane
at a
temperature of between 50 C and reflux or higher temperatures using microwave

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
28
irradiation.
Compounds of formula (TB) and formula (XIII) can also be prepared according to

Scheme C below from compounds of formula (VIII):
Scheme C
OtBu
Br Bu3SnOEt
N N Th\l)N1 N
---N ----N I
0 N Me 0 N Me I 0 N Me
Pd(PPh3)2C12
1M HCI(aq)
R3 DMF R3 R,
(V00 (X) (XI)
B R2
CN
B
R R2 2
NH2 N
Pv-_-D 1)1 Az-D N NN \
(XII)
IPA, cHCI 0 N Me ON Me
j'=-=
V
R3 R3
(IB) (XIII)
Compounds of formula (X) may be obtained from compounds of formula (VIII) by
reacting with tributy1(1-ethoxyvinyl)tin in the presence of a suitable
catalyst, for example
bis(triphenylphosphine)palladium(II) dichloride, in a suitable solvent such as
DMF
containing an aqueous acid such as 1M hydrochloric at a suitable temperature
between
ambient and the boiling point of the solvent.
Compounds of formula (XI) may be obtained from compounds of formula (X) by
reacting with a reagent such as Bredereck's reagent (tert-Butoxy-
bis(dimethylamino)methane) or dimethylformamide-dimethylacetal at a suitable
temperature between ambient and the boiling point of the solvent.
Compounds of formula (TB) and formula (XIII) may be obtained from compounds
of formula (XI) by reacting with compounds of formula (XII) in a solvent such
as IPA in
the presence of an acid such as concentrated hydrochloric acid.
Compounds of formula (II), formula (XXII) and formula ()MIT) can also be

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
29
prepared according to Scheme D below from compounds of formula (VIII):
Scheme D
B R2
R2
D
N
(IXX) Br AD
N
0 N Me (II)
B2(OH)4, KOAc
X-Phos, X-Phos-PdC12
K2CO3, IMS XR
B R2
BrB11:2
0 N Me (XX) Br
\ ¨NI (XXII)
0 N Me
R, B2(OH)4, KOAc
VIII) X-Phos, X-Phos-PdC12
(
K2CO3 , IMS
R2
B R2
)=N AzD N
¨N
(XXI) Br
0 N Me (XXIII)
B2(OH)4, KOAc
X-Phos, X-Phos-PdC12
K2CO3, IMS R,
Compounds of formulae (II), (XXII) and (XXIII) may be obtained from compounds
of formula (VIII) by reacting with compounds of formulae (IXX), (XX) and (XXI)
respectively in the presence of tetrahydroxydiboron, potassium acetate,
potassium
carbonate and a suitable catalyst, for example (2-dicyclohexylphosphino-
2',4',6'-
triisopropyl- 1 , 1 '-biphenyl)palladium(II) dichloride (X-Pho s-Pd-
C12) with 2-
dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-Phos), in a suitable
solvent such as
IMS at a suitable temperature between ambient and the boiling point of the
solvent.
These compounds of formulae (II), (XXII) and (XXIII) can be manipulated
further
to compounds of formula (I) using analogous procedures to those shown in
scheme A.

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
General Experimental Details
Reactions were not carried out under an inert atmosphere unless specified and
all
solvents and commercial reagents were used as received.
Purification by chromatography refers to purification using the CombiFlash
5
Companion purification system or the Biotage SP1 purification system. Where
products
were purified using an 'solute SPE Si II cartridge, `Isolute SPE Si
cartridge' refers to a
pre-packed polypropylene column containing unbonded activated silica with
irregular
particles with average size of 50 m and nominal 60A porosity. Fractions
containing the
required product (identified by TLC and/or LCMS analysis) were pooled and
concentrated
10 in
vacuo. Where HPLC was used for purification (Purification by MDAP) fractions
containing the required product (identified by TLC and/or LCMS analysis) were
pooled,
the bulk of the organic fraction removed by evaporation, and the remaining
aqueous
fraction lyophilised, to give the final product. Alternatively the pooled
product fraction was
evaporated to dryness under reduced pressure. Where thin layer chromatography
(TLC) has
15 been
used, it refers to silica gel TLC using plates, typically 3 x 6 cm silica gel
on aluminium
foil plates with a fluorescent indicator (254 nm), (e.g. Fluka 60778).
Microwave
experiments were carried out using a Biotage Initiator 6OTM which uses a
single-mode
resonator and dynamic field tuning. Temperature from 40-250 C can be achieved,
and
pressures of up to 30 bar can be reached.
20 NMR
spectra were obtained on a Varian Unity Inova 400 spectrometer with a 5 mm
inverse detection triple resonance probe operating at 400 MHz or on a Bruker
Avance DRX
400 spectrometer with a 5 mm inverse detection triple resonance TXI probe
operating at 400
MHz or on a Bruker Avance DPX 300 spectrometer with a standard 5mm dual
frequency
probe operating at 300 MHz. Shifts are given in ppm relative to
tetramethylsilane.
25 Compound names were generated using ACD/Name 2012.
Analytical LC-MS Conditions
LC-MS Method 1
The Waters ZQ quadrupole mass spectrometer with a Cl 8-reverse-phase column

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
31
(30 x 4.6 mm Phenomenex Luna 3 p.m particle size), elution with A: water +
0.1% formic
acid; B: MeCN + 0.1% formic acid. Gradient:
Gradient - Time flow (mL/min) %A %B
0.00 2.0 95 5
0.30 2.0 95 5
4.30 2.0 5 95
5.30 2.0 5 95
5.80 2.0 95 5
6.00 2.0 95 5
Detection - MS, UV (200 1/min split to the ESI source with in-line HP1100 PDA
detector)
MS ionisation method - Electrospray (positive and negative ion)
LC-MS Method 2
Waters Micromass ZMD quadrupole mass spectrometer with a C 18-reverse-phase
column (30 x 4.6 mm Phenomenex Luna 3 p.m particle size), elution with A:
water + 0.1%
formic acid; B: MeCN + 0.1% formic acid. Gradient:
Gradient - Time flow(mL/min) %A %B
0.00 2.0 95 5
0.50 2.0 95 5
4.50 2.0 5 95
5.50 2.0 5 95
6.00 2.0 95 5
Detection - MS, ELS, UV (200 pl split to MS with in-line UV detector)
MS ionisation method - Electrospray (positive and negative ion)
LC-MS Method 3
Waters Micromass ZQ2000 mass spectrometer with a C 18-reverse-phase column
(100 x 2.1 mm Acquity BEH with 1.7 jim particle size) maintained at 40 C,
elution with
A: water + 0.1% formic acid; B: MeCN + 0.1% formic acid. Alternatively, where
specified,

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
32
a C18-reverse-phase (100 x 2.1 mm Acquity UPLC BEH Shield 1.7 pm particle
size)
column was used.
Gradient:
Gradient ¨ Time flow (mL/min) %A %B
0.00 0.4 95 5
0.40 0.4 95 5
6.00 0.4 5 95
6.80 0.4 5 95
7.00 0.4 95 5
8.00 0.4 95 5
Detection - MS, UV PDA
MS ionization method - Electrospray (positive/negative ion).
LC-MS Method 4
Waters Platform LC quadrupole mass spectrometer with a Cl 8-reverse-phase
column (30 x 4.6 mm Phenomenex Luna 3 pm particle size), elution with A: water
+ 0.1%
formic acid; B: MeCN + 0.1% formic acid. Gradient:
Gradient ¨ Time flow (mL/min) %A %B
0.00 2.0 95 5
0.50 2.0 95 5
4.50 2.0 5 95
5.50 2.0 5 95
6.00 2.0 95 5
Detection - MS, UV (Split - 2000/min split to the ESI source with in-line
HP1100
DAD detection)
MS ionization method - Electrospray (positive and negative ion).
LC-MS Method 5
Waters VG Platform II quadrupole spectrometer with a C18-reverse-phase column
(30 x 4.6 mm Luna 3 pm particle size), elution with A: water + 0.1% formic
acid; B: MeCN

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
33
+ 0.1% formic acid.
Gradient:
Gradient - Time flow %A %B
0.00 2.0 95 5
0.30 2.0 95 5
4.30 2.0 5 95
5.30 2.0 5 95
5.80 2.0 95 5
6.00 2.0 95 5
Detection - MS, UV (Split ¨200 pl/min split to the ESI source with in-line
HP1050
DAD detection)
MS ionization method - Electrospray (positive and negative ion)
LC-MS Method U2
Acquity H-Class (quaternary pump/PDA detector) plus QDa Mass Spectrometer
with an Acquity UPLC BEH C18-reverse-phase column (1.7p.m particle size, 50 x
2.1mm
at 40 C), elution with A: water + 0.1% formic acid; B: MeCN + 0.1% formic
acid. Gradient:
Gradient ¨ Time flow (mL/min) %A %B
0.00 1.0 97 3
1.50 1.0 1 99
1.90 1.0 1 99
2.00 1.0 97 3
2.50 1.0 97 3
Detection - MS, UV
MS ionization method - Electrospray (positive and negative ion).

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
34
MDAP System:
Instrumentation: Agilent 1260 infinity purifications system. Agilent 6100
series
single Quadrupole LC/MS
Column: XSELECT CSH Prep C18 5 p.m OBD, 30X150 mm, RT
Mobile Phase A: 0.1% aqueous formic acid
Mobile Phase B: 0.1% formic acid in acetonitrile
Flow: 60 ml/min
Gradient Program: 10%-95%, 22 min, centred around a specific focused
gradient
Sample Injection of a 20-60mg/m1 solution in DMSO (+ optional formic acid and
water).
Abbreviations used in the experimental section:
AIBN Azobisisobutyronitrile
DCM Dichloromethane
DIPEA Di-isopropylethylamine
DMF N,N-dimethylformamide
DMSO Dimethylsulphoxide
Et20 Diethyl ether
Et0Ac Ethyl acetate
Hour
HPLC High performance liquid chromatography
IMS Industrial methylated spirits
LC-MS Liquid chromatography-mass spectrometry
MeCN Acetonitrile
MDAP Mass Directed Automatic Purification
Min Minutes
NBS N-Bromosuccinimide
Rt Retention time
RT Room temperature
THF Tetrahydrofuran

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
In the procedures that follow, some of the starting materials are identified
through
an "Intermediate" or "Example" number. The starting material may not
necessarily have
been prepared from the batch referred to.
When reference is made to the use of a "similar" or "analogous" procedure, as
5 will be appreciated by those skilled in the art, such a procedure may
involve minor
variations, for example reaction temperature, reagent/solvent amount, reaction
time,
work-up conditions or chromatographic purification conditions.
The invention will now be further described by the following examples.
Example 1
NC
N
0 N Me
C F3
4-(5-{5-Methyl-2-oxo-1-[3-(trifluoromethyl)pheny11-2,3-dihydro-1H-imidazol-
4-y11-1H-pyrazol-1-y1)benzonitrile
Intermediate 1
H N
NHO
411
CF3
1-Prop-2-yn-1-y1-3-13-(trifluoromethyl)phenyllurea
A solution of propargyl amine (4.41 g, 5.13 mL, 80 mmol) in MeCN (30 mL) was
added to a stirred solution of 3-(trifluoromethyl)phenyl isocyanate (15.0g,
12.12 mL,
80 mmol) in MeCN (60 mL) under a nitrogen atmosphere. The reaction mixture was
cooled
with a RT cooling bath and the rate of addition was such that the internal
temperature did
not exceed 35 C. After 1.5 h the mixture was concentrated in vacuo. Et0Ac (10
ml) was

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
36
added to the residue and the mixture was sonicated for 2 minutes. The
resultant slurry was
diluted with cyclohexane (40 ml). The mixture was stirred for 10 minutes and
the solid was
then recovered by filtration. The mother liquors were concentrated in vacuo
and the residue
dissolved in Et0Ac (10 mL). Dilution with cyclohexane (90 ml) precipitated a
second batch
of product which was recovered by filtration. The two batches were combined as
an ethyl
acetate solution and concentrated in vacuo to afford the title compound as a
fawn solid
(16.65 g).
LCMS (Method 2): Rt =3.22 min, mh 243 [M+Hr
Intermediate 2
ii-\11 \
CF3
1411111
1 0
5-Methyl-1-[3-(trifluoromethyl)phenyll-1,3-dihydro-2H-imidazol-2-one
A solution of 1-prop-2-yn-l-y1-343-(trifluoromethyl)phenyllurea (intermediate
1)
(11.2 g, 46 mmol) in THF (60 mL) and acetonitrile (120 mL) was added, under a
nitrogen
atmosphere, to a stirred suspension of sodium hydride (60% dispersion in
mineral oil)
(4.62 g, 115 mmol) in THF (60 mL) at such a rate that gas evolution was not
over-vigorous
and the internal temperature remained below 30 C. The mixture was stirred at
RT for 2.5
h, a thick precipitate having formed within 1 h. The reaction mixture was
cautiously
quenched with water (15 mL) and the resulting solution was treated with 1 M
hydrochloric
acid (150 mL, 150 mmol). The mixture was stirred for 4 hours then allowed to
stand for 15
hours. Saturated brine (150 mL) was added and the phases were partitioned. The
aqueous
phase was extracted with Et0Ac (100 mL). The combined organic phase was washed
with
saturated brine (100 mL), dried (sodium sulfate) and concentrated in vacuo.
The residue
was triturated with Et0Ac (33 mL). The resultant solid was taken into DCM and
filtered.
The filtrate was concentrated in vacuo to afford the title compound (10.0 g)
as a fawn solid.
LCMS (Method 1): Rt =2.63 min, mh 243 [M+H]

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
37
Intermediate 3
N--\\
----N
0 N Me
el CF3
2-Ethoxy-5-methyl-1-[3-(trifluoromethybphenyll-1H-imidazole
A solution of triethyloxonium tetrafluoroborate (9.0 g, 47 mmol) in DCM (62
mL)
was added to a stirred solution of 5-methyl-I -[3-(trifluoromethyl)phenyl]-1,3-
dihydro-2H-
imidazol-2-one (9.0 g, 37 mmol) in DCM (124 mL) under a nitrogen atmosphere.
The
solution was stirred at RT for 2.5 h then treated with water (50 mL) then 1 M
sodium
hydroxide (50 mL). The phases were partitioned. The aqueous phase was washed
with
DCM (2 x 50 mL). The combined organic phase was dried (sodium sulfate). The
solution
of the crude product was filtered through 2x50 g flash SCX 2 cartridges. Each
cartridge
was rinsed with 10% methanol in DCM (100 mL) then the product fraction was
eluted with
2M methanolic ammonia solution (100 mL). The fractions recovered with
methanolic
ammonia were combined and concentrated in vacuo to afford the title compound
(7.92 g)
as a brown solid.
LCMS (Method 1): Rt =2.41 min, m/z 271 [M+H]
Intermediate 4
Br
N\
----N _..1-..._
0 N Me
101 CF3
4-Bromo-2-ethoxy-5-methyl-1- [34trifluoromethyl)pheny11-1H-imidazole
A solution of NBS (5.16 g, 29 mmol) in MeCN (60 mL) was added to a stirred
solution of 2-ethoxy-5-methyl-143-(trifluoromethyl)pheny1]-1H-imidazole (7.92
g,
29 mmol) in MeCN (115 mL) at such a rate that the internal temperature of the
mixture did
not exceed 25 C (a RT cooling bath was used). After 0.5 h the mixture was
diluted with

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
38
water (50 mL) and saturated sodium carbonate (aq) (50 mL). Ethyl acetate (50
mL) was
added. The mixture was stirred vigorously then the phases were separated. The
organic
phase was washed with saturated brine (50 mL), dried (sodium sulfate) and
concentrated in
vacuo. The residue was taken into dichloromethane and filtered through a 25 g
Si II SPE
cartridge. The cartridge was washed with DCM and 10% Et0Ac in DCM. The
filtrate was
concentrated in vacuo to afford the title compound as an off-white solid (8.93
g).
LCMS (Method 1): Rt =3.86 min, m/z 349 [M(79Br) +H]
Intermediate 5
NC
N
N
0 N Me
CF3
445-{2-Ethoxy-5-methy1-1-13-(trifluoromethybphenyli-1H-imidazol-4-y1}-1H-
pyrazol-1-y1)benzonitrile
A solution of 4-bromo-2-ethoxy-5-methy1-1-[3-(trifluoromethyl)pheny1]-1H-
imidazole (3.14 g, 9 mmol) and 4-(5-tributylstannyl-pyrazol-1-y1)-benzonitrile
(W02014009425A1) (6.19 g, 13.5 mmol) in 1,4-dioxane (45 mL) was degassed by
bubbling argon through the solution for 5
minutes.
Tetrakis(triphenylphosphine)palladium(0) (0.52 g, 0.45 mmol) was added and
after a
further period of degassing the mixture was heated at 94 C under an argon
atmosphere
during 17 h. The mixture was cooled to RT and filtered through a 50 g flash
SCX 2
cartridge. The cartridge was eluted with DCM, 10% methanol in DCM then a 1:1
mixture
of 2M ammonia in methanol and DCM. Concentration of the appropriate fractions
in vacuo
afforded the crude product. This was triturated with cyclohexane and dried in
vacuo to
afford the title compound (3.01 g).

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
39
LCMS (Method 1): Rt =3.84 min, m/z 438 [M+H]
4-(5-{5-Methyl-2-oxo-1-[3-(trifluoromethyl)phenyll-2,3-dihydro-1H-imidazol-
4-y11-1H-pyrazol-1-y1)benzonitrile (Example 1)
A
suspension of 445- { 2-ethoxy-5-methy1-1 - [3 -(trifluoromethyl)phenyl] -1 H-
imidazol-4-y11-1H-pyrazol-1-yObenzonitrile (0.71 g, 1.6 mmol) in acetone (10
mL) and
1M hydrochloric acid (1.6 mL) was heated at 60 C for 14 h. The mixture was
cooled,
diluted with acetone (50 mL) and filtered through a 5 g flash NH2 column. The
filtrate was
concentrated in vacuo. The residue was twice taken into acetonitrile and re-
concentrated.
The residue was triturated with acetone (10 mL) and dried in vacuo to afford
the title
compound (0.45 g)
LCMS (Method 3): Rt=4.32 min, m/z 410.1 [M+H]
11-1 NMR (400 MHz, CDC13): 8 9.27 (111, br s), 7.78 (1H, d J=1.8 Hz), 7.73-
7.63
(4H, m), 7.60-7.56 (3H, m), 7.49 (1H, dJ=7.5 Hz), 6.56 (1H, d J=1.8 Hz), 1.84
(3H, S)
Example 2
NC = N-N
Me02S---\___\
0 N Me
C F3
4-(5- {5-Methyl-3-[3-(methylsulfonyl)propyll-2-oxo-1-E3-
(trifluoromethyl)phenyll-2,3-dihydro-1H-imidazol-4-y11-1H-pyrazol-1-
y1)benzonitrile
A mixture of 4-(5- {2-ethoxy-5-methy1-143-(trifluoromethyl)phenyl]-1H-imidazol-

4-yll-1H-pyrazol-1-yl)benzonitrile (intermediate 5) (66 mg, 0.15 mmol) and 1-
bromo-3-
methanesulfonyl propane (201 mg, 1.0 mmol) in acetonitrile (0.20 mL) was
heated at 120 C
for 6 h. The mix was cooled to RT and diluted with acetonitrile (2 mL) and
water (1 mL)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
then treated with 1M sodium hydroxide (0.20 m1).The mixture was heated at 50 C
for 2 h.
The cold mixture was diluted with water (10 mL) and extracted with Et0Ac (2 x
10 mL).
The combined organic phase was dried (sodium sulfate) and concentrated in
vacuo. The
residue was purified by MDAP to afford the title compound (10 mg).
5 LCMS (Method 3): Rt=4.38 min, m/z 530.1 [M+H]
1H NMR (400 MHz, CDC13): 6 7.89 (1H, d J=1.8 Hz), 7.74 (2H, d J=8.8 Hz), 7.71-
7.57 (511, m), 7.51 (1H, d J=7.7 Hz), 6.69 (1H, d J=1.8 Hz), 3.86-3.74 (1H,
m), 3.42-3.32
(1H, m), 3.04 (211, t J=8.0 Hz), 2.87 (3H, s), 2.10-1.96 (2H, m), 1.75 (3H, s)
Example 3
NC
-N
Me.
N
\
ONN Me
CF3
4-(5-13,5-Dimethy1-2-oxo-1-13-(trifluoromethyl)pheny11-2,3-dihydro-1H-
imidazol-4-y11-1H-pyrazol-1-y1)benzonitrile
Sodium hydride (10 mg of a 60% dispersion in mineral oil, 0.25 mmol) was added
to a stirred suspension of 4-(5-{5-methy1-2-oxo-1-[3-(trifluoromethyl)pheny1]-
2,3-
dihydro-1H-imidazol-4-y11-1H-pyrazol-1-y1)benzonitrile (Example 1) (70 mg,
0.17 mmol)
in THF (2.0 mL) under a nitrogen atmosphere. This resulted in degassing and
the formation
of a yellow solution. Iodomethane (0.031 mL, 0.5 mmol) was added and the
mixture was
stirred for 3 h. Water (10 mL) was added and the mixture was extracted with
DCM (2 x 10
mL). The combined organic phase was dried (sodium sulfate) and concentrated in
vacuo.
The residue was purified by flash column chromatography (2 g Si II SPE eluted
with 0-
50% Et0Ac in DCM in 10 % increments). Concentration of the appropriate
fractions in
vacuo gave the title compound which was freeze-dried from acetonitrile/water
to afford a

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
41
fluffy white solid (50 mg).
LCMS (Method 3): Rt=4.57 min, m/z 424.1 [M+11]
1H NMR (400 MHz, CDC13): 8 7.87 (111, d J=1.8 Hz), 7.74 (214, d J=8.7 Hz),
7.69-
7.57 (5 Hz, m), 7.51 (111, d J=7.7 Hz), 6.62 (1H, d J=1.8 Hz), 2.98 (3H, s),
1.76 (3H, s)
Example 4
NC la,
RIP -N
I 10
N-4
0 N Me
CF3
Ethy1-5-11-(4-eyanopheny1)-1H-pyrazol-5-y11-4-methy1-2-oxo-3-1-3-
(trifluoromethyl)phenyll-2,3-dihydro4H-imidazole-1-earboxylate
Sodium hydride (38 mg of a 60% dispersion in mineral oil, 0. 95 mmol) was
added
to a solution of 4-(5- {5-methy1-2-oxo-1- [3 -(trifluoromethyl)phenyl] -
2,3-dihydro-1H-
imidazol-4-y1 -1H-pyrazol-1-yebenzonitrile (Example 1) (300 mg, 0.73 mmol) in
DMF
under a nitrogen atmosphere. The mixture was stirred for 5 minutes and the
resultant yellow
solution was treated with ethyl chloroformate (104 mg, 0.091 mL, 0.95 mmol).
The mixture
was stirred for 1 h then treated with water and extracted with Et0Ac. The
organic phase
was washed with water and brine then dried (sodium sulfate) and concentrated
in vacuo.
The residue was purified by chromatography using silica gel eluting with 25%-
50% ethyl
acetate in cyclohexane. The product thus obtained was freeze-dried from
acetonitrile-water
to afford the title compound (194 mg) a white solid.
LCMS (Method 3): Rt=4.83 min, m/z 482.1 [M+H]
1H NMR (400 MHz, CDC13): 67.82 (1H, d J=1.7 Hz), 7.76-7.60 (7H, m), 7.52 (1H,
d J= 7.9 Hz), 6.56 (1H, d J=1.7 Hz), 4.17-4.07 (2H, m), 1.84(311, s),
1.12(311, t J=7.1 Hz)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
42
Example 5
NC
-N
Me
0 N
0 N Me
CF
445-{3-Acetyl-5-methyl-2-oxo-1-l3-(trifluoromethyl)phenyll-2,3-dihydro4H-
imidazol-4-y11-1H-pyrazol-1-y1)benzonitri1e
Sodium hydride (12.5 mg of a 60% dispersion in mineral oil, 0.31 mmol) was
added
to a
stirred suspension of 4-(5- {5-methy1-2-oxo-1-[3-(trifluoromethypphenyl]-2,3-
dihydro-1H-imidazol-4-y1 -1H-pyrazol-1-yl)benzonitrile (Example 1) (102 mg,
0.25 mmol) in THF (5.0 mL) under a nitrogen atmosphere. This resulted in
degassing and
the formation of a yellow solution. Acetyl chloride (0.022 mL, 0.5 mmol) was
added and
the mixture was stirred for 1 h then left to stand for 16 h. Water (10 mL) was
added and the
mixture was extracted with Et0Ac (2 x 10 mL). The combined organic phase was
dried
(sodium sulfate) and concentrated in vacuo. The residue was purified by flash
column
chromatography (2 g Sill SPE eluted with DCM then 10% Et0Ac in DCM).
Concentration
of the appropriate fractions in vacuo gave the title compound which was freeze-
dried from
acetonitrile/water to afford a fluffy white solid (56 mg).
LCMS (Method 3): Rt=4.86 mm, iniz 452.0 [M+H]+
NMR (400 MHz, CDC13) 8 7.80 (11-1, d J=1.7 Hz), 7.77-7.66 (4H, m), 7.63-7.51
(4H, m), 6.51 (1H, d J=1.7 Hz), 2.44 (3H, s), 1.88 (3H, s)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
43
Example
NC
40 NN
0
H i \
0--;-N Me
4111 C F3
541-(4-Cyanopheny1)-1H-pyrazol-5-yll-N-ethyl-4-methyl-2-oxo-3-13-
(trifluoromethyl)pheny11-2,3-dihydro-1H-imidazole-1-earboxamide
4-Nitrophenyl chloroformate (226mg, 1.12 mmol) was added portion wise to a
solution of 4-(5- {5-methy1-2-oxo-143-(trifluoromethyl)pheny1]-2,3-dihydro-1H-
imidazol-
4-y11-1H-pyrazol-1-yObenzonitrile (Example 1) (409 mg, 1.0 mmol) and
triethylamine
(0.355 mL, 2.55 mmol) in DCM (12 mL) under a nitrogen atmosphere. The mixture
was
stirred for 2.75 h giving a yellow solution. A solution of ethylamine (3.0 mL
of a 2M
solution in THF, 6 mmol) was added. The mixture was stirred for a further 2.5
h then
concentrated in vacuo. The residue was chromatographed on silica gel eluting
with 10-40%
Et0Ac in DCM then re-columned on silica gel with 25-60% Et0Ac in cyclohexane
to
afford the title compound as a colourless foam after drying in vacuo.
LCMS (Method 3): Rt=4.95 mm, m/z 481.1 [M+Hr
1H NMR (400 MHz, CDC13): 6 8.38 (1H, t J=5.3 Hz), 7.81 (1H, d J=1.8 Hz), 7.77-
7.66 (4H, m), 7.63-7.58 (3H, m), 7.53 (1H, d J=7.9 Hz), 6.56 (1H, d J=1.8 Hz),
3.21-3.13
(2H, m), 1.87 (3H, s), 1.04 (3H, t J=7.2 Hz).
In Table 1 Method A refers to compounds prepared in an analogous manner to
example 4, Method B refers to compounds prepared in analogous manner to
example 5,
Method C refers to compounds prepared in analogous manner to example 6 and
Method D
refers to compounds prepared in an analogous manner to example 3 (where salts
of amines
were used in place of the free base additional triethylamine was added to the
reaction
mixture to allow for this). In the table below, where rotameric signals have
been identified

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
44
in the NMR spectrum, these have been labelled by *.
Table 1
Ex Structure Method 1H NMR LC-MS
Method 3
7 NC
-N 'H NMR (400 MHz, Rt=4.36
S
CDC13): 8 8.74, (1H, min, m/z
0
t J=5.5 Hz), 7.81 =497.1
(1H, d J=1.8 Hz), [M+H]
7.78-7.58 (7H, m),
H(3,-N Me .1 7.53 (1H, d =7.9
N
Hz), 6.56 (1H, d
CF J=1.8 Hz), 3.61 (2H,
q J=5.4 Hz), 3.33
3
(2H, q, J=5.4 Hz),
541-(4-Cyanopheny1)-1H- 1.88 (3H, s), 1.79
pyrazol-5-y1]-N-(2- (1H, t J=5.3 Hz)
hydroxyethyl)-4-methy1-2-
oxo-343-
(trifluoromethyl)pheny1]-2,3-
dihydro-1H-imidazole-1-
carboxamide
8 NC1HNMR (400 MHz, Rt=4.42
OH 141111 N DMS0): 8 8.36 (1H, min, m/z=
0 m-
t J=5.7 Hz), 8.01 511.1
(1H, s), 7.94-7.78 [M+H]+
(6H, m), 7.66 (2H, d
H 0"-' N \ Me J=8.7Hz), 6.63 (1H,
d J=1.8 Hz), 4.41
CF (1H, t J=4.9 Hz),
3.36-3.20 (2H, m,
3
partially obscured by
5-[1-(4-Cyanopheny1)-1H- water peak), 3.0 (2H,
pyrazol-5-y1]-N-(3- q J=6.3Hz), (1.88,
hydroxypropy1)-4-methyl-2- 3H, s), 1.41-1.23
oxo-343- (2H, m)
(trifluoromethyl)pheny1]-2,3-
dihydro-1H-imidazole-l-
carboxamide
(continued)

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
9 NC C 1H NMR (400 MHz, Rt=3.51
NCDC13): 8 8.57 (111, min, m/z---
0 m-N
br s), 7.81 (1H, d 538.2
J=1.7 Hz), 7.77-7.66 [M+H]
NAN
(411, m), 7.63-7.59
H
0 NMe
(3H, m), 7.75 (1H, d
J=7.5 Hz), 6.55 (1H,
CF d J=1.7 Hz), 3.24-
3.16 (2H, m), 2.34
(2H, t J=7.3 Hz),
5-[1-(4-Cyanopheny1)-1H- 2.26 (6H, s), 1.87
pyrazol-5-yli-N-[3- (311 s), 1.66-1.58
(dimethylamino)propy11-4- (2H, m)
methy1-2-oxo-343-
(trifluoromethyl)phenyl]-2,3-
dihydro-1H-imidazole-1-
carboxamide
gal
m-N 1H NMR (400 MHz, Rt=5.28
10 NC
CDC13): 1H NMR min, m/z=
0 (400 MHz, CDC13) 8 480.1
7.80 (1H, d J=1.8 [M+H]
N Hz), 7.76-7.51 (8H,
ON-Me m), 6.51 (1H, d
J=1.8 Hz), 2.89 (1H,
4111) CF d J=17.4 Hz of t
J=7.2 Hz), 2.75 (111,
3
d J= 17.4 Hz of t
4-(5-{3-Butanoy1-5-methy1-2- J=7.4 Hz), 1.90 (3H,
oxo4-[3- s), 1.49-1.39 (211,
(trifluoromethyl)pheny1]-2,3- m), 0.79 (311, t J=7.4
dihydro-1H-imidazol-4-yll- Hz)
1H-pyrazol-1-yl)benzonitrile
11 NC light 1H NMR (400 MHz, Rt=4.84
,N
CDC13): 8 8.65 (111, min, m/z=
s), 7.81 (1H, d J=1.6 511.1
Hz), 7.77-7.59 (7H, [M+H]
N N m), 7.53 (1H, d
H0 J=7.8 Hz), 6.55 (111,
N Me
d J=1.6 Hz), 3.42-
1411111 C F3 3.29 (7H, m), 1.85
(3H, s)
5-[1-(4-Cyanopheny1)-1H-
pyrazol-5-y1]-N-(2-
methoxyethyl)-4-methyl-2-
oxo-3-[3-
(trifluoromethyl)pheny1]-2,3-
dihydro-1H-imidazole-1-
carboxamide
(continued)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
46
12 NC
1 I, C IH NMR (400 MHz, Rt=3.47
N N-N CDC13): 8 8.60 (1H, min, m/z=
.1
t J=4.9 Hz), 7.81 524.1
CNIN-------) (1H, d J=1.8 Hz), [M+H]
H.., \ BA_ 7.76-7.59 (711, m),
ON N 1"'
7.52 (1H, d J=7.9
elCF, Hz), 6.55 (1H, d
J=1.8 Hz), 3.23 (2H,
5-[1-(4-Cyanopheny1)-1H- q J=5.8 Hz), 2.32
pyrazol-5-y1]-N-[2- (2H, t J=6.2 Hz),
(dimethylamino)ethy1]-4- 2.18 (6H, s), 1.84
methyl-2-oxo-3[3- (3H, s)
(trifluoromethyl)pheny1]-2,3-
dihydro-1H-imidazole-1-
carboxamide
13 NC C IH NMR (400 MHz, Rt=4.52
me 10 CDC13): d 8.99 (1H, mm,
0=sI 0 fN t J=5.8 Hz), 7.83 miz=559.0
di
NAN µ ---- (1H, d J=1.8 Hz), [M+H]+
7.77-7.65 (4H, m),
0-----'pN me 7.63-7.59 (3H, m),
7.53 (111, d J=8.0
S cF, Hz), 6.56 (1H, d
J=1.8 Hz), 3.76-3.62
5-[1-(4-Cyanopheny1)-1H-
(2H, m), 3.10 (2H, t
pyrazol-5-y1]-4-methyl-N-[2-
J=6.2 Hz), 2.90 (3H,
(methylsulfonypethy1]-2-oxo-
s), 1.85 (3H, s)
3-[3-
(trifluoromethyl)pheny1]-2,3-
dihydro-1H-imidazole-l-
carboxamide
14 NC
WI _N C Ill NMR (400 MHz, Rt=4.76
CDC13): 8 8.65 (1H, mm,
a
ID
_1.1 j
, t J=6.0 Hz), 7.82 m/z=523.1
o AN
(1H, d J=1.8 Hz), [M+H]+
N
H \ 7.78-7.66 (4H, m),
0 N Me
7.63-7.57(311, m),
S CF 7.53 (111, d J=7.6
Hz), 6.57-6.55 (1H,
,
m), 4.26-4.16 (111,
5-[1-(4-Cyanopheny1)-1H- m), 3.89-3.72 (3H,
pyrazol-5-y1]-4-methy1-2- m), 3.63 (0.5H*, m),
oxo-N-(tetrahydrofuran-3-y1)- 3.40 (0.5 H*, m),
3-[3- 2.22-2.07 (111, m),
(trifluoromethyl)pheny1]-2,3- 1.89 (3H, s), 1.81-
dihydro-1H-imidazole-1- 1.72 (0.5H*, m),
carboxamide 1.64-1.56 (0.511*, m)
(continued)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
47
15 NC VI gel
m ., N C 1H NMR (400 MHz, Rt=4.76
CDC13): 8 8.65 (1H, t min,
0
......)
J=6.0 Hz), 7.82 (1H, d m/z=523.1
N N
J=1.8 Hz), 7.78-7.66 [M+H]+
,
H 0-_,/ \ me (4H, m), 7.63-7.57
-.:---'N
(311, m), 7.53 (1H, d
101 CF, J=7.6 Hz), 6.57-6.55
(111, m), 4.26-4.16
(1H, m), 3.89-3.72
5-[1-(4-Cyanopheny1)-1H- (3H, m), 3.63 (0.511*,
pyrazol-5-y1]-4-methyl-2- m), 3.40 (0.5 H*, m),
oxo-N-[(3S)-tetrahydrfuran- 2.22-2.07 (1H, m),
1.89 (311, s), 1.81-1.72
(trifluoromethyl)pheny1]-2,3- (0.5H*, m), 1.64-1.56
dihydro-1H-imidazole-1- (0.5H*, m)
carboxamide
16 NC gli _a_t
Ki - N C 1FINMR (400 MHz, Rt=4.72
CDC13): 8 8.65 (1H, t min,
OaNo - \ J=6.0 Hz), 7.82 (1H, d
m/z=523.2
AN--) J=1.8 Hz), 7.78-7.66 [M+H]+
H dN \ (411, m), 7.63-7.57
0 N Me
(3H, m), 7.53 (11I, d
1. CF J=7.6 Hz), 6.57-6.55
(1H, m), 4.26-4.16
,
(111, m), 3.89-3.72
5-[1-(4-Cyanopheny1)-1H- (3H, m), 3.63 (0.5H*,
pyrazol-5-y1]-4-methyl-2- m), 3.40 (0.5 11*, m),
oxo-N-[(3R)-tetrahydrofuran- 2.22-2.07 (1H, m),
1.89 (311, s), 1.81-1.72
(trifluoromethyl)pheny1]-2,3- (0.5H*, m), 1.64-1.56
dihydro-1H-imidazole-1- (0.5H*, m)
carboxamide
17 NC .4µt
4111 -N C 1H NMR (400 MHz, Rt=4.69
CDC13): 8 8.83 (1H, min,
o t J=5.2Hz), 7.81 (1H, m/z=547.1
ellH _C\ d J=1.7 Hz),7.76- [M+H]+
pr-N 0----'-"N MeP 7.49 (811, m), 7.25
Me (111, d J=2.2 Hz)
0
CF3 6.56 (1H, d J=1.7
Hz), 5.91 (111, d
5-[1-(4-Cyanopheny1)-1H- J=2.2 Hz), 4.37 (1H,
pyrazol-5-y1]-4-methyl-N-[(1- d J=15.0 Hz of d
methyl-1H-pyrazol-3- J=5.7 Hz), 4.27 (111,
yl)methy1]-2-oxo-3-[3- d J=15.0 Hz of d
(trifluoromethyl)pheny1]-2,3- J=4.9), 3.83 (311, s),
dihydro-1H-imidazole-1- 1.87 (3H, s)
carboxamide
(continued)

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
48
18 NC riet
-N A 1H NMR (400 MHz, Rt=4.63
CDC13): 6 7.82 (1H, mm,
0
d J=1.8 Hz), 7.76- nilz=468.1
7.70 (3H, m), 7.69- [M+H]+
N 7.59 (4H, m), 7.51
0 N Me (1H, d J=7.9 Hz),
6.57 (1H, d J=1.8
C F3 Hz), 3.71 (3H, s),
1.84 (3H, s)
Methy1-5-[1-(4-
cyanopheny1)-1H-pyrazol-5-
y1]-4-methy1-2-oxo-3-[3-
(trifluoromethyl)phenyl]-2,3-
dihydro-1H-imidazole-1-
carboxylate
19 NC
-N 1H NMR (400 MHz, Rt=5.46
CDC13): 8 7.80 (1H, min,
0 d J=1.7 Hz), 7.76- m/z=494.1
7.65 (4H, m), 7.64- [M+H]+
\AN \ 7.51 (4H, m), 6.50
(1H, d J=1.7 Hz),
0 N Me
2.95-2.74 (2H, m),
CF 1.90 (3H, s), 1.42-
1.32 (2H, m), 1.22-
1.12 (2H, m), 0.83
445- 5-Methy1-2-oxo-3 - (3H, t J=7.4 Hz)
pentanoy1-143-
(trifluoromethyl)pheny1]-2,3-
dihydro-1H-imidazol-4-y11-
1H-pyrazol-1-y1)benzonitrile
20 NC gal
N-N 1H NMR (400 MHz, Rt=5.23
CDC13): 6 7.84 (1H, mm, m/z
d J=1.8 Hz) 7,81- 524.2
o
7.73 (4H, m), 7.68- [M+Na]+
7.58 (2H, m), 7.55
0 /
(1H, s), 7.44 (1H, d
ON Me
J=7.8 Hz), 7.57 (1H,
14101 CF3 d J=1.8 Hz), 4.40-
4.20 (2H, m), 1.48
(3H, s), 1.37 (9H, s)
tert-Butyl- {5-[1-(4-
cyanopheny1)-1H-pyrazol-5-
y1]-4-methy1-2-oxo-3-[3-
(trifluoromethyl)phenyl]-2,3-
dihydro-1H-imidazol-1-
ylf acetate

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
49
Example 21
NC la,
m-N
1
N-(
I
N \)
N Me
CF3
541-(4-Cyanopheny1)-1H-pyrazol-5-yll-N-ethyl-N,4-dimethyl-2-oxo-3-13-
(trifluoromethyl)pheny11-2,3-dihydro-1H-imidazole-1-carboxamide
4-(5- 5-Methyl-2-oxo-1 - [3 -(trifluoromethyl)phenyl] -2,3 -dihydro-1H-
imidazol-4-
y11-1H-pyrazol-1-yObenzonitrile (Example 1) (107 mg, 0.262 mmol) was stirred
in DCM
(3 mL) and triethylamine (0.109 mL, 0.786 mmol) with N,N-dimethylaminopyridine

(2 mg). To this mixture was added 4-nitrophenyl chloroformate (106mg, 0.524
mmol) and
the resulting mixture stirred at RT for 1 h. N-methylethylamine (93 mg, 1.57
mmol) in
DCM (1 mL) was then added and stirring was continued for a further 2 h. The
mixture was
diluted with more DCM, washed with water, the organic phase separated, dried
over
Na2SO4 and evaporated. The product was purified by chromatography on a 12 g
silica gel
column, eluting with a gradient of 20% to 60% Et0Ac in cyclohexane. The gum
thus
obtained was dissolved in a small quantity (< 1 mL) of Et0Ac and diluted with
cyclohexane
resulting in a formation of a white solid which was filtered off and dried.
(65 mg, 50%).
LCMS (Method 3): Rt=4.70 min, m/z 495.1 [M+H]
1H NMR (400 MHz, DMSO at 80 C): 8 7.93 (211, d J=8.4 Hz), 7.83 (1H, d J=1.8
Hz), 7.81-7.72 (5H, m), 7.68-7.64 (1H, m), 6.62 (114, d J=1.8 Hz), 3.24 (2H, q
J=6.7 Hz),
2.80 (3H, s), 1.63 (3H, s), 1.0 (3H, t J=6.8 Hz).

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
Example 22
NC ig6
m-N
HO,
inN
0
0 N Me
CF3
{5-[1-(4-Cyanopheny1)-1H-pyrazol-5-y11-4-methyl-2-oxo-313-
(trifluoromethyl)pheny11-2,3-dihydro-1H-imidazol-1-vlIacetic acid
5 tert-Butyl- {5- [1-(4-cyanopheny1)-1H-pyrazol-5-yl] -4-methyl-2-oxo -3 -
[3 -
(trifluoromethyl)phenyl] -2,3-dihydro-1H-imidazol-1-y1 acetate (Example 20)
(75 mg,
0.143 mmol) was stirred in DCM (3 mL) and water (0.03 mL). To this mixture was
added
TFA (0.1 mL) and the mixture stirred overnight. Reaction was incomplete so
further TFA
(0.5 mL) was added and after lh 30 min. a further 1 ml was added. After 4 h
reaction was
10 complete, solvents were evaporated and the residue partitioned between
Et0Ac and water.
The organic phase was separated, dried over Na2SO4 and evaporated to give the
title
compound as a gum.
LCMS (Method 1): Rt 3.14 min., [M+H] 468
1H NMR (CDC13): 8 7.87 (1H, d J=1.8Hz), 7.75-7.66 (5H, m), 7.61 (1H, t J=8.0
15 Hz), 7.55 (1H, s), 7.43 (1H, d J=8.0 Hz), 6.63 (1H, d J=1.8 Hz), 4.46-
4.32 (2H, m), 1.50
(3H, s) one proton not observed.
Example 23
NC ail
11µ qjj No-NI
¨N
o
0 N Me
14I C F3

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
51
2-{5-[144-Cyanopheny1)-1H-pyrazol-5-y11-4-methyl-2-oxo-3-13-
(trifluoromethyl)phenyll-2,3-dihydro-1H-imidazol-1-yll-N-methylacetamide
1,1'-carbonyl diimidazole (45 mg, 0.278 mmol) was added to a solution of {541-
(4-cyanopheny1)-1H-pyrazol-5-yl] -4-methyl-2-oxo-3-[3-(trifluoromethyl)phenyl]
-2,3-
dihydro-1H-imidazol-1-yll acetic acid (Example 22) (65 mg, 0.139 mmol) in THF
(2.0 mL). The mixture was stirred for 1.25 h then a 2M methylamine in THF
(0.278 mL,
0.556 mmol) was added. The mixture was stirred for 45 min then a second
aliquot of 2M
methylamine in THF (0.278 mL, 0.556 mmol) was added. The mixture was stirred
for a
further 40 minutes then partitioned between Et0Ac and water. The phases were
partitioned.
The organic phase was dried over Na2SO4 and evaporated. The product was
purified by
chromatography on a 12 g silica gel column, eluting with 5% methanol in DCM.
The
material thus obtained was freeze dried from acetonitrile-water to afford the
title compound
(35 mg) a white solid.
LCMS (Method 3): Rt=4.10 mm, m/z 481.1 [M+H]
1H NMR (400 MHz, CDC13): 8 7.86 (1H, d J=1.8 Hz), 7.76-7.60 (6H, m) 7.56, (1H,
s) 7.45 (1H, d J=7.7Hz), 6.68 (111, d J=1.8 Hz), 6.23 (1H, hr s), 4.28-4.08
(2H, m), 2.75
(3H, d J=4.8 Hz), 1.56 (3H, s overlapping with water signal).
Example 24
NiC 0
NA0 -N
1.......)
NI k
9 ON Me
S.
0 8'0
14111
CF3

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
52
Benzenesulfonate 3- [({5-11-(4-cyanopheny1)-1H-pyrazol-5-yli -4-methyl-2-oxo-
3- [3-(trifluoromethyl)pheny11-2,3-dihydro-1H-imidazol-1-yllearbonyl)aminol-
N,N,N-trimethylpropan-l-aminium
Methyl benzenesulfonate (0.019 mL, 0.14 mmol) was added to a solution of 5-[1-
(4-
cyanopheny1)-1H-pyrazol-5-y1]-N43-(dimethylamino)propy1]-4-methy1-2-oxo-343-
(trifluoromethyl)pheny1]-2,3-dihydro-1H-imidazole-1-carboxamide (Example 9)
(77 mg,
0.14 mmol) in THF (3.0 mL). The mixture was stirred for 2hr then allowed to
stand at RT for
2 days. Water (15 mL) and Et0Ac (15 ml) were added with stirring. The phases
were separated.
The aqueous phase was washed with Et0Ac (15 mL) then diluted with MeCN. The
solvents
were removed by lyophilisation to afford the title compound (70 mg) a white
solid.
LCMS (Method 3): Rt=3.52 min, m/z 552.2 [Mr
1H NMR (400 MHz, DMS0): 8 8.47 (1H, t J=6.0 Hz) 7.99-7.80 (7H, m), 7.69 (2H,
d J=8.7 Hz) 7.61-7.58 (2H, m), 7.34-7.28 (3H, m), 6.64 (1H, d J=1.8 Hz) 3.15
(2H, t J=8.2
Hz), 3.04-2.97 (11H, m), 1.89 (3H, s), 1.73-1.63 (2H, m)
The following compound was prepared by analogous procedure to that used in
Example 24.
Table 2
Ex Structure 1H NMR LC-MS
Method
3
1H NMR (400 MHz, Rt=3.51 min
NC
I DMS0): 8 8.61 (1H, t m/z=538.1
,õ,,N J=5.9 Hz) 8.02-7.82 (711, [M]+
m) 7.70 (2H, d J=8.8
Hz), 7.61-7.57 (211, m),
1:)N Me
S- 7.34-7.26 (3H, m), 6.65
8-0 at
(1H, d J=1.7 Hz) 3.45-
W CF, 3.35 (2H, m), 3.22-
Benzenesulfonate 2-[({5-[1-(4- 3.10(2H, m), 2.99 (9H,
cyanopheny1)-1H-pyrazol-5-y1]-4- s), 1.90 ( 3H, s)
methy1-2-oxo-343-
(trifluoromethyl)pheny1]-2,3-
dihydro-1H-imidazol-1-
yll carbonyl)amino]-N,N,N-
trimethylethanaminium

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
53
Example 26
NC
-N
N¨c\
Co N Me
14D CF3
445- {3-Butyl-5-methyl-2-oxo-1- [3-(trifluoromethyl)phenyll-2,3-dihydro-1H-
imidazo1-4-01-1H-pyrazo1-1-yl)benzonitrile
445- {5-Methyl-2-oxo-143-(trifluoromethyl)phenyl] -2,3 -dihydro-1H-imidazol-4-
y11-1H-pyrazol-1-y1)benzonitrile (Example 1) (102 mg, 0.25 mmol) was added to
a stirred
mixture of potassium tert-butoxide (42 mg, 0.375 mmol) and THF (2.0 mL) under
a
nitrogen atmosphere. This resulted in the formation of a yellow solution. 1-
Iodo-butane
(0.057 mL, 0.5 mmol) was added and the mixture was stirred for 7 h then left
to stand for
16 h. Water (10 mL) was added and the mixture was extracted with DCM (3x10
mL). The
combined organic phase was dried (sodium sulfate) and concentrated in vacuo.
The residue
was purified by flash column chromatography (2 g Si II SPE eluted with DCM).
The crude
product thus obtained was further purified by flash column chromatography (2 g
Si IT SPE
eluted with 20% Et0Ac in cyclohexane). Concentration of the appropriate
fractions in
vacuo gave the title compound which was freeze-dried from acetonitrile/water
to afford a
white solid (25 mg).
LCMS (Method 3): Rt=5.24 min, m/z 466.1 [M+H]
1H NMR (400 MHz, CDC13): 1H NMR (400 MHz, CDC13) 6 7.87 (1H, d J=1.7 Hz),
7.73 (2H, d J=8.7 Hz), 7.67- 7.57 (5H, m), 7.50 (1H, d J=7.5 Hz), 6.63(11I d
J=1.7 Hz),
3.61 (1H, m), 3.20 (1H, m) 1.72 (3H, s), 1.50-1.35 (2H, m), 1.27-1.15, (2H,
m), 0.81 (3H,
t J=7.4 Hz)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
54
Example 27
NC
m-N
0õ0
Me,S,N
0 N Me
CF3
4-(5-{5-Methyl-3-(methylsulfony1)-2-oxo-1-[3-(trifluoromethyl)pheny11-2,3-
dihydro-1H-imidazol-4-01-1H-pyrazol-1-y1)benzmitrile
Sodium hydride (12 mg of a 60% dispersion in mineral oil, 0.30 mmol) was added

to a solution of 4-(5-{5-methy1-2-oxo-1-[3-(trifluoromethyl)phenyl]-2,3-
dihydro-1H-
imidazol-4-y11-1H-pyrazol-1-yObenzonitrile (Example 1) (94 mg, 0.23 mmol) in
DMF
(1.5 mL) under a nitrogen atmosphere. The mixture was stirred for 5 minutes
and the
resultant yellow solution was treated with methanesulfonyl chloride (32 mg,
21.4 L,
0.28 mmol). The mixture was stirred for 2 h then treated with water and
extracted with
Et0Ac. The organic phase was washed with water and brine then dried (sodium
sulfate)
and concentrated in vacuo. The residue was purified by chromatography using
silica gel
eluting with 25%-60% ethyl acetate in cyclohexane. The product thus obtained
was
freeze-dried from acetonitrile-water to afford the title compound (54 mg) a
white solid.
LCMS (Method 3): Rt=4.70 min, m/z= 488.1 [M+H]
1H NMR (400 MHz, CDC13): 1H NMR (400 MHz, CDC13) 6 7.83 (1H, d J=1.8 Hz),
7.80-7.73 (3H, m), 7.69 (1H, t J=7.9 Hz), 7.66 (3H, m), 7.56 (1H, d J=8.0 Hz),
6.63 (1H, d
J=1.8 Hz), 3.07 (3H, s), 1.92 (3H, s)
The following compounds were prepared by analogous procedure to that used in
Example 26. In the table below, where rotameric signals have been identified
in the NMR
spectrum, these have been labelled by *.

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
Table 3
Ex Structure 1H NMR LC-MS
Method
3
28 NC & 1H NMR (400 MHz, d6- Rt = 5.49 min,
m-N DMS0): 8 8.04-7.98 m/z = 480.1
(3H, m), 7.85-7.66 (6H, [M+H]+
CA\ Me m), 6.83-6.77 (1H*, m),
N
3.45-3.12 (1H*, m),
CF3 1.87-1.52 (4H*, m),
1.33-1.22 (1H, m), 1.08-
4-{5-[5-Methy1-3-(1-methyl-buty1)- 0.79(5H*, m), 0.69-0.65
2-oxo-1-(3-trifluoromethyl- (3H*, m)
pheny1)-2,3-dihydro-1H-imidazol-
4-yl]pyrazol-1-y1}-benzonitrile
29 NC 1H NMR (400 MHz, d6- Rt = 3.37 min,
-N DMS0): 5 7.97-7.94 m/z = 481.2
ji (3H, m), 7.78-7.69 (5H, [M+H]+
CAN"---Me m), 7.60 (1H, d, J=7.6
Hz), 6.86 (1H, d, J=1.8
CF3 Hz), 3.62-3.54 (1H, m),
3.45-3.42 (1H, m), 2.29-
4- { 5- [3 -(2-Dimethylamino-ethyl)- 2.22 (1H, m), 2.16-2.10
5-methyl-2-oxo-1-(3- (1H, m), 1.88 (6H, s),
trifluoromethyl-phenyl)-2,3- 1.46 (3H, s)
dihydro-1H-imidazol-4-y1]-pyrazol-
1-yll-benzonitrile
30 NC
-N 1H NMR (400 MHz, d6- Rt = 3.41 min,
nf
DMS0): 5 8.03-7.97 m/z = 495.3
i
(3H, m), 7.83-7.78 (2H, [M+H]+
N m), 7.76 (1H, t, J=8 Hz),
o ,
7.72-7.65 (3H, m), 6.87
CF, (1H, d, J=1.83 Hz), 3.53-
3.38 (1H, m), 3.20-3.07
4- {5- [3 -(3 -Dimethylamino-propy1)- (1H, m), 2.10-2.00 (2H,
5-methyl-2-oxo-1-(3- m), 1.97 (6H, s), 1.68
trifluoromethyl-phenyl)-2,3- (3H, s), 1.57-1.42 (111,
dihydro-1H-imidazol-4-y1]-pyrazol- m), 1.41-1.26 (1H, m)
1-y1}-benzonitrile
The following compounds were prepared by analogous procedure to that used in
Example 27.
5

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
56
Table 4
Ex Structure 1H NMR LC-MS
Method
3
31 NC 1H NMR (400 MHz, Rt = 4.98 mm,
CDC13): 8 7.83 (1H, d, miz = 516.2
o o
J=1.7 Hz), 7.78-7.71 [M+H]+
ON\ Me (3H, m), 7.69-7.60 (4H,
m), 7.55 (1H, dm, J=7.9
CF, Hz), 6.61 (1H, d, J=1.8
Hz), 3.51 (1H, ddd, J=14,
4- {5-[5-Methy1-2-oxo-3-(propane- 9, and 6 Hz), 3.11 (1H,
1-sulfony1)-1-(3-trifluoromethyl- ddd, J=14, 9, and 6 Hz),
phenyl)-2,3-dihydro-1H-imidazol- 1.89 (3H, s), 1.71-1.57
4-y1]-pyrazol-1-yll-benzonitrile (2H, m), 0.96 (3H, t,
J=7.5 Hz)
32 NC op 1H NMR (400 MHz, d6- Rt = 4.87 min,
o P N-N DMS0): 8 8.03-
7.95 miz = 514.2
(3H, m), 7.93-7.84 (3H, [M+H]+
V oN\ m), 7.83 (1H, tm,
J=7.7Hz), 7.75-7.68 (2H,
CF, m), 6.78 (111, d, J=1.75
Hz), 2.99 (1H, m), 1.87
4-{5[3-Cyclopropanesulfony1-5- (3H, s), 1.12-0.93 (3H,
methyl-2-oxo-1-(3-trifluoromethyl- m), 0.81-0.73 (1H, m),
phenyl)-2,3-dihydro-1H-imidazol- no NH observed
4-y1]-pyrazol-1-y1 -benzonitrile
33 NC grab 1H NMR (400 MHz, d6- Rt = 4.86 min,
41,1 ,N DMS0): 8 8.51 (1H, t, miz = 517.2
o o =
J=5.7 Hz), 7.96-7.92 [M+H]+
H \ (3H, m), 7.87-7.84 (2H,
oN me
m), 7.80-7.79 (2H, m),
OP CF, 7.68-7.64 (2H, m), 6.69
(1H, d, J=1.8 Hz), 2.59-
5-[2-(4-Cyano-pheny1)-2H-pyrazol- 2.42 (2H, m), 1.85 (3H,
3-y1]-4-methyl-2-oxo-3-(3- s), 0.84 (3H, t, J=7.3 Hz)
trifluoromethyl-pheny1)-2,3-
dihydro-imidazole-1-sulfonic acid
ethylamide
(continued)

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
57
34 NC
N.N DMNMR (400 MHz, d6- Rt = 5.17 min,
S0): 8 8.65 (1H, d, miz = 557.2
J=7.9 Hz), 8.01-7.95 [M+H]+
N N
Hod, N \ me (2H, m), 7.93 (1H, m),
7.92-7.87 (2H, m), 7.87-
1410CF3 7.78 (2H, m), 7.73-7.66
(2H, m), 6.71 (1H, d,
5-[2-(4-Cyano-phenyl)-2H-pyrazol- J=1.75 Hz), 2.79 (1H,
3-y1]-4-methyl-2-oxo-3-(3- m), 1.93 (3H, s), 1.60-
trifluoromethyl-pheny1)-2,3- 1.43 (4H, m), 1.42-1.16
dihydro-imidazole-l-sulfonic acid (4H, m)
cyclopentylamide
35 NC lab NMR
(400 MHz, d6- Rt = 4.85 mm,
DMS0): 5 8.95 (1H, bs), miz = 529.1
A'Ng:g9- 8.00-7.93 (3H, m), 7.92- [M+H]+
1-10.2 A 7.86 (2H, m), 7.85-7.78
N me
(2H, m), 7.75-7.68 (2H,
CF3 m), 6.71 (1H, d, J=1.77
Hz), 2.16 (1H, m), 1.84
5-[2-(4-Cyano-phenyl)-2H-pyrazol- (3H, s), 0.54-0.37 (3H,
3-y1]-4-methy1-2-oxo-3-(3- m), 0.37-0.28 (1H, m)
trifluoromethyl-pheny1)-2,3-
dihydro-imidazole-1-sulfonic acid
cyclopropylamide
The following compounds were prepared by analogous procedure to that used in
Example 23. In the table below, where rotameric signals have been identified
in the NMR
spectrum, these have been labelled by *.
Table 5
Ex Structure 'H NMR LC-MS
Method 3
36 NC 40 'FINMR (400 MHz, d6- Rt = 4.43 mm,
m-N
DMS0): 8 8.02-7.97 m/z = 521.2
)1--\ N (2H, m), 7.96-7.90 (3H, [M+H]+
o\ m), 7.81-7.71 (2H, m),
oN me
7.65 (1H, m), 7.55 (1H,
CF3 dm, J=7.7 Hz), 6.79 (1H,
d, J=1.8 Hz), 4.44 (2H,
4- {5-[5-Methy1-2-oxo-3-(2-oxo-2- s), 3.41-3.35 (2H, m),
pyrrolidin-1-yl-ethyl)-1-(3- 3.24-3.09 (2H, m), 1.88-
trifluoromethyl-pheny1)-2,3- 1.79 (2H, m), 1.74-1.65
dihydro-1H-imidazol-4-yThpyrazol- (2H, m), 1.29 (3H, s)
1-yll-benzonitrile
(continued)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
58
37 NC 416 N \ 'FT NMR (400 MHz, d6- Rt = 4.27 mm,
VI -N DMS0): 8 8.01-7.99 m/z = 495.2
(2H, m), 7.94-7.91 (3H, [M+H]+
Me

111
ocAN\ ), 7.81-7.71 (2H, m),
7.65 (1H, m), 7.58 (111,
0 CF, dm, J=7.8 Hz), 6.79 (111,
d, J=1.9 Hz), 4.62-4.51
2-[5-[2-(4-Cyano-phenyl)-2H- (211, m), 2.97 (3H, s),
pyrazol-3-y1]-4-methyl-2-oxo-3-(3_ 2.76 (311, s), 1.31 (3H, s)
trifluoromethyl-pheny1)-2,3-
dihydro-imidazol-l-yl] -N,N-
dimethyl-acetamide
38 NC
\
N W,N 'II NMR (400 MHz, d6- Rt = 3.33 mm,
.4., DMS0): 8 8.03-7.97 m/z = 550.2
(211, m), 7.95 (1H, d, [M+H]+
ood,N\ me J=1.8 Hz), 7.92-7.88
(211, m), 7.81-7.72 (211,
0 CF3 m), 7.66 (111, m), 7.58
(1H, d, J=7.7 Hz), 6.78
4-{5-[5-Methyl-3-[2-(4-methyl- (111, d, J=1.8 Hz), 4.66-
piperazin-1-y1)-2-oxo-ethyl]-2-oxo- 4.52 (211, m), 3.46-3.39
1-(3-trifluoromethyl-pheny1)-2,3- (211, m), 3.39-3.32 (211,
dihydro-1H-imidazol-4-y11-pyrazol- m), 2.23-2.23 (211, m),
1-yll-benzonitrile 2.23-2.07 (2H, m), 2.16
(311, s), 1.32 (311, s)
39 NC
'II NMR (400 MHz, d6- Rt = 3.24 mm,
.--N DMS0): 8 8.15 (1H, s), nilz = 552.3
.11._).` 8.08 (111, t, J=5.5 Hz), [M+11p-
0 2 1,--"\"
cr'-i'N \ Me 7.99-7.93 (2H, m), 7.90
HO j (1H, d, J=1.8 Hz), 7.88-
011111 c3 7.82 (2H, m), 7.78-7.68
(2H, m), 7.64 (1H, m),
2-[542-(4-Cyano-pheny1)-2H- 7.56 (111, dm, J=7.8 Hz),
pyrazol-3-y1]-4-methyl-2-oxo-3-(3- 6.75 (111, d, J=1.8 Hz),
trifluoromethyl-phenyl)-2,3- 4.264.10 (2H, m), 3.01-
dihydro-imidazol-1-y1]-N-(3- 2.91 (2H, m), 2.19-2.12
dimethylamino-propy1)-acetamide (211, m), 2.09 (611, s),
formic acid salt 1.45-1.36 (211, m), 1.32
(311, s), salt NH not
observed
(continued)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
59
, 111 NMR (400 MHz, d6- Rt = 4.80 min,
40 NC
DMS0): 8 8.03-7.97 m/z = 535.4
,
0.¨Nr\N (2H, m), 7.94-7.87 (3H, [M+II]+
d,\ m), 7.78 (1H, dm, J=7.8
o N me
Hz), 7.74 (111, tm,
40 CF, J=7.8), 7.65 (1H, bs),
7.58 (1H, dm, J=7.7),
2-[5-[2-(4-Cyano-phenyl)-2H- 6.78 (0.5H*, d, J=1.5
pyrazol-3-y1]-4-methy1-2-oxo-3-(3- Hz), 6.76 (0.5H*, d,
trifluoromethyl-phenyl)-2,3- J=1.5 Hz), 4.71-4.27
dihydro-imidazol-1-y1]-N- (3H, m), 2.91 (1.5H*, s),
cyclobutyl-N-methyl-acetamide 2.74 (1.5H*, s), 2.27-
1.79 (4H, m), 1.69-1.49
(2H, m), 1.33 and 1.31 (
3H*, 2 singlets)
The following compounds were prepared by analogous procedure to that used in
Example 6. Where salts of amines were used in place of the free base,
additional triethyl amine
was added to the reaction mixture to allow for this. In the table below, where
rotameric signals
have been identified in the NMR spectrum, these have been labelled by *.
Table 6
Ex Structure 1H NMR LC-MS
Method
3
41 NC
-N 'El NMR (400 MHz, d6- Rt = 5.24 min,
DMS0): 8 8.37 (1H, d, m/z = 507.2
N
J=7.6 Hz), 7.97 (1H, s), [M+H]+
HcAN \ me 7.89-7.76 (6H, m), 7.62-
7.59 (2H, m), 6.60 (1H,
CF3 d, J=1.8 Hz), 3.83 (1H,
sext. J=7.8 Hz), 2.06-
5-[2-(4-Cyano-pheny1)-2H-pyrazol- 1.90 (2H, m), 1.84 (3H,
3-y1]-4-methyl-2-oxo-3-(3- s), 1.81-1.73 (1H, m),
trifluoromethyl-phenyl)-2,3- 1.68-1.46 (3H, m)
dihydro-imidazole-l-carboxylic
acid cyclobutylamide
(continued)

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
42 NC A II NMR (400 MHz, d6- Rt = 4.95 mm,
WI -N DMS0): 6 8.32 (1H, d, m/z = 493.2
&N-i:Lnif-e J=3.1 Hz), 7.98 (1H, bs), [M+H]+
H \ 7.94-7.87 (3H, m), 7.86
0 N Me (1H, d, J=1.75 Hz), 7.85-
*CF3 7.78 (2H, m), 7.86 (1H,
d, J=1.75 Hz), 7.85-7.78
5-[2-(4-Cyano-phenyl)-2H-pyrazol- (2H, m), 7.69-7.64 (211,
3-y1]-4-methyl-2-oxo-3-(3- m), 6.34 (1H, d, J=1.75
trifluoromethyl-phenyl)-2,3- Hz), 2.38 (1H, octet),
dihydro-imidazole-l-carboxylic 0.63-0.49 (2H, m), 0.39-
acid cyclopropylamide 0.28 (1H, m), 0.16-0.05
(1H, m)
43 NC la, 'II NMR (400 MHz, d6- Rt = 4.93 min
RP
3
\--%: DMS0): 8 d 8.50 m/z = 628.3
(0.51-1*, d, J=7.18 Hz), [M+H]+
NO'NIN N ji---- 8.41 (0.5H*, d, J=7.6
Hod,--NL.
Hz), 8.03 (111, m), 7.95-
*CF3 7.79 (6H, m), 7.69-7.61
(2H, m), 6.64 (1H, d,
5-[2-(4-Cyano-pheny1)-2H-pyrazol- J=1.75 Hz), 3.56-3.39
3-y1]-4-methy1-2-oxo-3-(3- (111, m), 3.27-2.83 (6H,
trifluoromethyl-phenyl)-2,3- m), 1.91 and 1.90 (311*,
dihydro-imidazole-l-carboxylic 2 x s), 1.69-1.33 (4H, m),
acid (1-ethanesulfonyl-piperidin-4- 1.23 (1.511*, t, J=7.3 Hz),
y1)-amide 1.15 (1.511*, t, J=7.3 Hz).
44 NC rat
WI -N 111 NMR (400 MHz, d6- Rt = 5.83 mm,
DMS0): 6 8.30 (111, d, miz = 537.2
---...,
NjN-- ---) J=7.2 Hz), 8.02 (1H, m), [M+H]+, 559.2
L\N
H 7.94-7.90 (2H,m), 7.90- (M+Na)+
0 N\ Me 7.87 (111, m), 7.87-7.82
0CF3 (3H,m ), 7.68-7.62 (21-1,
m), 6.64 (1H, bs), 3.74-
5-[2-(4-Cyano-pheny1)-2H-pyrazol- 3.59 (2H, m), 3.52-3.40
3-y1]-4-methy1-2-oxo-3-(3- (111, m), 3.32-3.22 (2H,
trifluoromethyl-phenyl)-2,3- m), 1.90 (3H, s), 1.68-
dihydro-imidazole-1-carboxylic 1.57 (111, m), 1.43-1.26
acid (tetrahydro-pyran-4-y1)-amide (2H, m), 1.20-1.03 (1H,
m)
(continued)
5

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
61
45 NC
glj NMR
(400 MHz, d6- Rt = 5.42 min,
MS0): 8 8.23 (1H, d, m/z = 521.2
9
NN D
J=6.7 Hz), 8.02 (1H, m), [M+H]+, 543.2
7.94-7.89 (2H,m), 7.89- (M+Na)+
ii 7.94-7.89 \ me
7.78 (4H, m), 7.67-7.62
CF, (2H, m), 6.64 (11-1, bs),
3.67 (1H, m), 1.90 (3H,
5-[2-(4-Cyano-phenyl)-2H-pyrazol- s), 1.78-1.63 (1H, m),
3-y1]-4-methyl-2-oxo-3-(3- 1.60-1.38 (5H, m), 1.35-
trifluoromethyl-pheny1)-2,3- 1.19 (111, m), 1.08-0.94
dihydro-imidazole-l-carboxylic (1H, m)
acid cyclopentylamide
46 NC rah. '11 NMR (400 MHz, d6- Rt = 3.49 min,
DMS0): 8 8.41 (1H*, 2 m/z = 536.2
N 0
A x d, J=7 Hz), 8.02 (1H, [M+H]+
"N N m), 7.97-7.78 (6H,m),
Hod,N
7.68-7.61 (2H, m), 6.64
(1H*, 2 x s), 3.83 (1H,
m), 2.62-2.49 (1H, m),
5-[2-(4-Cyano-pheny1)-2H-pyrazol- 2.42-1.99 (4H, m), 2.18
3-y1]-4-methyl-2-oxo-3-(3- (3H, s), 1.98-1.79 (4H,
trifluoromethyl-phenyl)-2,3- m)
dihydro-imidazole-l-carboxylic
acid ((R)-1-methyl-pyrrolidin-3-
y1)-amide
47 NC ah
411 -N '11NMR (400 MHz, d6- Rt =3.49 min,
No
DMS0): 8 8.41 (1H*, 2 m/z = 536.2
elaA
N 0 N ---- x d, J=7.2 Hz), 8.02 (1H, [M+H]+
m), 7.96-7.78 (6H,m),
7.68-7.62 (2H, m), 6.64
0110 cF3 (1H*, 2 x s), 3.83 (1H,
bs), 2.62-2.49 (1H, m),
542-(4-Cyano-pheny1)-2H-pyrazol- 2.41-2.00 (4H, m), 2.18
3-y1]-4-methyl-2-oxo-3-(3- (3H, s), 1.96-1.80 (4H,
trifluoromethyl-phenyl)-2,3- m)
dihydro-imidazole-l-carboxylic
acid ((S)-1-methyl-pyrrolidin-3-y1)-
amide
(continued)

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
62
48 NC ati
-N 1H NMR (400 MHz, d6- Rt = 4.38 min,
DMS0): 8 8.45-8.32 m/z = 539.2
N
(1H, m), 8.01 (1H, m), [M+H]+
HO Flo\ 7.97-7.72 (6H, m), 7.71-
N 7.62 (2H, m), 6.65 (1H,
CF3 d, J=1.76), 5.30 (1H, m),
3.94 (1H, bs), 3.62-3.38
Mixture of 542-(4-Cyano-phenyl)- (5H, m), 1.89 (3H, bs),
2H-pyrazol-3-y1]-4-methyl-2-oxo- signals broadened due to
3-(3-trifluoromethyl-pheny1)-2,3- presence of rotamers
dihydro-imidazole-l-carboxylic
acid ((3R,4S)-4-hydroxy-
tetrahydro-furan-3-y1)-amide
and
5-[2-(4-Cyano-pheny1)-2H-pyrazol-
3-y1]-4-methy1-2-oxo-3-(3-
trifluoromethyl-pheny1)-2,3-
dihydro-imidazole-1-carboxylic
acid ((3S,4R)-4-hydroxy-
tetrahydro-furan-3-y1)-amide
49 NC cal
-N 1H NMR (400 MHz, d6- Rt = 4.30 min,
N
DMS0): 8 9.06 (1H, t, m/z = 544.2
0
HcA\ J=6 Hz), 8.45 (11-1, ddd, [M+H]+
J=4.9, 1.7 and 1 Hz),
N
8.04 (111, m), 7.94-7.80
1410 C F3 (6H, m), 7.69 (1H, td,
J=7.8 and 1.8 Hz), 7.67-
5-[2-(4-Cyano-pheny1)-2H-pyrazol- 7.62 (2H, m), 7.26 (111,
3-y1]-4-methyl-2-oxo-3-(3- ddd, J=7.8, 4.9, and 1
trifluoromethyl-phenyl)-2,3- Hz), 6.98 (1H, dm, J=7.8
dihydro-imidazole-l-carboxylic Hz), 6.64 (1H, d, J=1.76
acid (pyridin-2-ylmethyl)-amide Hz), 4.27 (2H, m), 1.89
(3H, s)
50 NC aei
-N 1H NMR (400 MHz, d6- Rt = 3.84 min,
DMS0): 6 d 8.89 (1H, t, m/z = 544.2
HcA \ J=6 Hz), 8.44 (1H, dd, [M+H]+
J=4.9, 1.7 Hz), 8.28 (1H,
N
dm, J1.9 Hz), 8.03 (1H,
c3 m), 7.92-7.79 (6H, m),
7.65-7.59 (2H, m), 7.33
542-(4-Cyano-pheny1)-2H-pyrazol- (1H, dt,1=8 and 1.9 Hz),
3-y1]-4-methyl-2-oxo-3-(3- 7.27 (1H, ddd, J=8, 4.7,
trffluoromethyl-phenyl)-2,3- and 0.8 Hz), 6.63 (1H, d,
dihydro-imidazole-l-carboxylic J=1.76 Hz), 4.30-4.06
acid (pyridin-3-ylmethyl)-amide (2H, m), 1.90 (3H, s)
(continued)

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
63
51 NC
N-N 11-1NMR (400 MHz, d6- Rt = 3.69 min,
DMS0): 6 8.91 (1H, t, miz = 544.2
J=6 Hz), 8.45-8.40 [M+H]+
r,1 H (2H,m), 8.05 (1H, bs),
0 N 7.93-7.87 (4H, m), 7.86-
CF, 7.80 (2H, m), 7.68-7.61
(211, m), 6.96-6.90 (2H,
5-[2-(4-Cyano-phenyl)-2H-pyrazol- m), 6.64 (111, d, J=1.76
3-y1]-4-methyl-2-oxo-3-(3- Hz), 4.30-4.09 (2H, m),
trifluoromethyl-phenyl)-2,3- 1.92 (3H, s)
dihydro-imidazole-l-carboxylic
acid (pyridin-4-ylmethyp-amide
52 NC a h
14 _ N 1H NMR (400 MHz, d6- Rt = 4.57 mm,
DMS0): 8 9.07 (111, t, m/z = 545.0
o J=5.7 Hz), 8.52 (2H, m), [M+H]+
= H 8.30 (111, m), 8.04 (1H,
0 N m), 7.93-7.80 (6H, m),
1410CF3 7.65-7.60 (211, m), 6.63
(1H, d, J=1.76 Hz), 4.42-
542-(4-Cyano-pheny1)-2H-pyrazol- 4.26 (211, m), 1.89 (311,
3-y1]-4-methyl-2-oxo-3-(3- s)
trifluoromethyl-pheny1)-2,3-
dihydro-imidazole-1-carboxylic
acid (pyrazin-2-ylmethyl)-amide
53 NC
1H NMR (400 MHz, d6- Rt = 4.68 min,
DMS0): 6 8.23 (111, d, miz = 537.1
J=6.5 Hz), 8.01 (111, m), [M+H]+
I-10 Hod,N 7.94-7.78 (6H, m), 7.69-
7.61 (2H, m), 6.64 (111,
CF3 d, J=1.76 Hz), 4.80-4.63
(111*, m), 3.75-3.64 (1H,
5-[2-(4-Cyano-pheny1)-2H-pyrazol- m), 3.47-3.38 (111*, m),
3-y1]-4-methyl-2-oxo-3-(3- 1.89 (311, s), 1.77-1.31
trifluoromethyl-phenyl)-2,3- (4H, m), 1.30-1.17 (111,
dihydro-imidazole-l-carboxylic m), 1.04-0.90 (1H, m)
acid ((1R,2R)-2-hydroxy-
cyclopenty1)-amide
54 NC gel
-N 1H NMR (400 MHz, d6- Rt = 3.56 min,
DMS0): 8 8.40 (1H, t, m/z = 564.1
J=6 Hz), 8.03 (1H, m), [M+H]+
N
N H 7.93-7.79 (6H, m), 7.68-
0 N
7.61 (211, m), 6.62(111,
CE, d, J=1.74 Hz), 2.97-2.87
(1H, m), 2.79-2.70 (111,
5-[2-(4-Cyano-phenyl)-2H-pyrazol- m), 2.69-2.62 (211, m),
3-y1]-4-methyl-2-oxo-3-(3- 2.11(311, s), 1.92 (3H, s),
trifluoromethyl-phenyl)-2,3- 1.76-1.62 (2H, m), 1.34-
dihydro-imidazole-1-carboxylic 1.16 (211, m), 1.16-1.02
acid (1-methyl-piperidin-4- (1H, m), 0.98-0.82 (2H,
ylmethyp-amide m)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
64
Example 55
00
NC s
K N
0 N Me
C F3
3-Methanesulfony1-4-{5-[5-methy1-2-oxo-1-(3-trifluoromethyl-pheny1)-2,3-
dihydro-1H-imidazol-4-y11-pyrazol-1-y11-benzonitrile
Intermediate 6
0 0
õ
NC s
-N
N
0 N Me
CF3
4-{5-[2-Ethoxy-5-methy1-1-(3-trifluoromethyl-pheny1)-1H-imidazol-4-y11-
pyrazol-1-y11-3-methanesulfonyl-benzonitri1e
In a vial were mixed 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (19
mg,
0.04 mmol) and (2-dicyclohexylphosphino-2 ',4',6'-triisopropy1-
1,1'-
biphenyl)palladium(II) dichloride (16mg, 0.02 mmol), tetrahydroxydiboron (270
mg, 3
mmol) and potassium acetate (294 mg, 3 mmol) and the vial flushed with N2. A
solution of
4-bromo-2-ethoxy-5-methy1-1- [3 -(trifluoromethyl)pheny1]-1H-imidazole
(Intermediate 4,
349 mg, 1 mmol) in IMS (10 ml) was added and the sealed vial heated at 80 C
for 1 h. A
degassed solution of K2CO3 (1.8M, 1.66 ml) was then added followed by a
solution of 4-
(5-bromo-pyrazol-1 -y1)-3 -methanesulfonyl-benzonitrile (DE102010030187,
Example 6A,
326 mg, 1 mmol) in THF (3 m1). Heating was continued for a further 2h. The
solution was
decanted from the insoluble inorganics and evaporated. The residue was
triturated with

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
Et0Ac and evaporated again to give an orange gum which was purified by silica
chromatography eluting with a gradient of 20% to 100% Et0Ac in cyclohexane.
Product
fractions were combined, evaporated, redissolved in MeCN-water and freeze
dried to give
Intermediate 6 as a white solid (40 mg).
5 LCMS (Method 2 Rt = 3.39 min., m/z 516.3 [M+H]+
3-Methanesulfony1-4-f5-[5-methyl-2-oxo-1-(3-trifluoromethyl-phenyl)-2,3-
dihydro-1H-imidazol-4-yll-pyrazol-1-y11-benzonitrile (Example 55)
The Intermediate 6 was heated in a mixture of acetone (5 ml) and 1M HC1 (2 ml)
at
60 C overnight. After cooling, the acetone and most of the water were removed
by
10
evaporation in vacuo and the concentrate was diluted with MeCN and water and
freeze
dried. The crude product (36 mg) was purified by reverse phase chromatography
(125 g
C18, eluant 10% to 90% MeCN-water containing 0.1% formic acid) to afford the
title
compound (16 mg).
LCMS (Method 3): Rt = 4.18 min, m/z 488.1 [M+H]+
15 NMR
(400 MHz, d6-DMS0): 6 10.65 (1H, s), 8.52 (1H, d, J=1.8 Hz), 8.35 (1H,
dd, J=8.3, 2.0 Hz), 7.93 (1H, d, J=2.0 Hz), 7.76-7.64 (3H, m), 7.65 (1H, d,
J=8.3 Hz), 7.56-
7.51 (1H, dm, J=7.8 Hz), 6.73 (1H, d, J=1.8 Hz), 3.59 (3H, s), 1.51 (3H, s).
Example 56
NC Br
=
NN
j)(
N N
H
0 N Me
20 CF3

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
66
5-[2-(2-Bromo-4-cyano-pheny1)-211-pyrazol-3-y111-4-methyl-2-oxo-343-
trifluoromethyl-pheny1)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide
Intermediate 7
0
0 N Me
C F3
1-[2-Ethoxy-5-methy1-1-(3-trifluoromethyl-phenyl)-1H-imidazol-4-y11-
ethanone
4-Bromo-2-ethoxy-5-methyl-1- [3 -(trifluoromethyl)phenyl] -1H-imidazole
(Intermediate 4, 700 mg, 2 mmol), tributy1(1-ethoxyvinyl)tin (940 mg, 2.6
mmol),
bis(triphenylphosphine)palladium(II) dichloride (70 mg, 0.1 mmol), DMF (7 ml)
and 1M
HC1 (14 ml) were mixed in each of 3 sealed, degassed (N2) vials which were
heated at
120 C for 1.5 h. The reaction mixtures were combined and treated with 1M HC1
(50 ml)
and stirred vigorously for 30 min. Water and Et0Ac were added and the
separated organic
phase was washed with water (x2) brine, dried (Na2SO4) filtered and evaporated
to give a
brown oil which was purified by chromatography using 10% to 40% Et0Ac in
cyclohexane
to give Intermediate 7 as a yellow oil which crystallized upon cooling (0.91
g, 34%).
LCMS (Method 2) Rt = 3.20 min., m/z 285.2 [M+H]+
Intermediate 8
0
N¨?1 µ-N/
Me
CF,
3-Dimethylamino-1-[2-ethoxy-5-methy1-1-(3-trifluoromethyl-pheny1)-1H-
imidazol-4-01-propenone
1- [2-Ethoxy-5-methy1-1 -(3-trifluoromethyl-phenyl)-1H-imidazol-4-yl] -
ethanone

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
67
(Intermediate 7, 1.68 g, 5.38 mmol) and Bredereck's reagent (8 ml) were mixed
and heated
at 100 C for a total of 3h then allowed to stand for 2 days. The Bredereck's
reagent was
partially evaporated in vacuo at 50 C and the concentrate was diluted with
Et0Ac, washed
with water (x3), brine, dried (Na2SO4) and evaporated to give Intermediate 8
as an orange
gum which crystallized on standing (1.8 g, 91%).
LCMS (Method 2) Rt = 2.85 min., m/z 368.3 [M+H]+
Intermediate 9
NC Br
N
0 N Me
CF3
3-Bromo-4-{5-1-5-methyl-2-oxo-143-trifluoromethyl-phenyl)-2,3-dihydro-1H-
imidazol-4-yll-pyrazol-1-yll-benzonitrile
3-Dimethylamino-1- [2-ethoxy-5-methy1-1 -(3 -trifluoromethyl-pheny1)-1H-
imidazol-
4-y1]-propenone (Intermediate 8, 100 mg, 0.27 mmol) was dissolved in isopropyl
alcohol (2
ml) and conc. hydrochloric acid (0.1 ml) was added followed by 3-bromo-4-
hydrazino-
benzonitrile (70 mg, 0.32 mmol). The solution was heated at 85 C for 2h.
Solvent was
removed in vacuo and the residue was purified by column chromatography using
0% to 100%
Et0Ac in cyclohexane to give Intermediate 9 as a pink solid (90 mg, 68%).
LCMS (Method U2) Rt = 1.39 min., m/z 488.1 and 490.0 [M+H]+ (Br isotopes)
5-1242-Bromo-4-cyano-phenyl)-2H-pyrazol-3-01-4-methyl-2-oxo-3-(3-
trifluoromethyl-phenyl)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide
(Example 56)
3-Bromo-4- { 5[5-methy1-2-oxo-1-(3 -trifluoromethyl-phenyl)-2,3 -dihydro -1H-
imidazol-4-yThpyrazol-1-yll -benzonitrile (Intermediate 9, 120 mg, 0.246
mmol),
cyclopentyl isocyanate (740, 0.74 mmol) and DIPEA (129 Ill, 0.74 mmol) were
stirred

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
68
together in DCM (5 ml) at room temperature for 4h then at 45 C overnight.
After cooling,
the solvent was removed and the product purified by silica gel chromatography
eluting with
0% to 50% Et0Ac-cyclohexane to afford the title compound as a colourless foam
(96mg,
65%).
LCMS (Method 3): Rt = 5.64 min, m/z 599.2 & 601.2 [M+H]+, m/z 621.2 & 623.2
[M+Na]+
1HNMR (400 MHz, CDC13): 8 8.36 - 8.32 (1H, d, J=7 Hz), 7.95 (1H, d, J=1.7 Hz),

7.83 (1H, d, J=1.8 Hz), 7.76 (1H, d, J=8.2 Hz), 7.73 (1H, dm, J=7.8 Hz), 7.68
(1H, dd,
J=8.2, 1.8 Hz), 7.66 (1H, t, J=7.8 Hz), 7.57-7.55 (111, m), 7.47 (1H, dm,
J=8.2 Hz), 6.55
(1H, d, J=1.7 Hz), 4.00 (1H, tdt, J=6.6, 6.4, 6.7 Hz), 1.93 (3H, s), 1.91-1.89
(2H, m), 1.68-
1.56 (4H, m), 1.41-1.30 (2H, m).
Example 57
//
I.
-N r
/
CLN 51( N
Me
CF,
5-11-(2-Bromo-4-cyano-pheny1)-1H-pyrazol-3-y11-4-methy1-2-oxo-3-(3-
trifluoromethyl-phenyl)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide
Intermediate 10
//
I.
Br
-N
I /
ICIN Me
CF,

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
69
3-Bromo-4-0-[5-methyl-2-oxo-1-(3-trifluoromethy1-pheny1)-2,3-dihydro4H-
imidazol-4-yll-pyrazol-1-y11-benzonitri1e
3 -Dimethylamino-1 - [2-ethoxy-5-methy1-1-(3-trifluoromethyl-pheny1)-1H-
imidazol-4-yl] -propenone (Intermediate 8, 540 mg, 1.47 mmol) and 3-bromo-4-
hydrazino-
benzonitrile (374 mg, 1.76 mmol) were heated together in IPA (15 ml) with
conc.
hydrochloric acid (5 drops) at 85 C for 3h. A further 5 drops of conc. HC1
were added and
heating was continued for a further 2h. Solvent was evaporated and the residue
was
chromatographed (0% to 100% Et0Ac-cyclohexane). The first eluting component
(31 mg,
4.3%) was Intermediate 10, followed by the 2nd eluting component Intermediate
9 (270
mg, 38%).
LCMS (Method 2) Rt = 3.28 min., m/z 488.1 and 490.1 (Br isotopes)
51142-Bromo-4-cyano-phenyl)-1H-pyrazol-3-y11-4-methyl-2-oxo-3-(3-
trifluoromethyl-phenyl)-2,3-dihydro-imidazole-l-carboxylic acid
cyclopentylamide
(Example 57)
Following the procedure described in Example 56 using cyclopentyl isocyanate
Intermediate 10 was converted to the title compound.
LCMS (Method 3): Rt = 5.88 min, m/z 599.2 and 601.2 [M+H]+, m/z 621.2 and
623.2 [M+Na]+
1H NMR (400 MHz, d6-DMS0): 68.62 (1H, d, J=7.1 Hz), 8.47 (1H, d, J=1.8 Hz),
8.30 (1H, d, J=2.6 Hz), 8.05 (1H, dd, J=8.3, 1.8 Hz), 7.93-7.91 (1H, m), 7.88-
7.76 (3H, m),
7.77 (1H, d, J=8.3 Hz), 6.59 (1H, d, J=2.6 Hz), 4.07-3.96 (1H, m), 2.04 (3H,
s), 1.91-1.81
(2H, m), 1.67-1.57 (2H, m), 1.56-1.46 (4H, m)
Example 58
NC F
0 NON
o
aNAN_,
H
N Me
CF,

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
5-12-(4-Cyano-2-fluoro-phenyl)-2H-pyrazol-3-y11-4-methy1-2-oxo-343-
trifluoromethyl-phenyl)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide
The title compound was synthesized from 3-Fluoro-4-hydrazino-benzonitrile and
Intermediate 8 using a similar procedure to that described in Example 56.
5 LCMS (Method 3): Rt = 5.52 min, m/z 539.2 [M+H]+, m/z 561.1 [M+Na]+
IFINMR (400 MHz, CDC13): 8 8.33 (1H, d, J=6.8 Hz), 7.85 (1H, d, J=1.8 Hz),
7.78-
7.72 (2H, m), 7.68 (1H, t, J=7.8 Hz), 7.59 (1H, m), 7.56 (1H, ddd, J=0.9, 1.7,
8.2 Hz), 7.51
(1H, dm, J=8.1 Hz), 7.47 (1H, dd, J= 9.7 and 1.7 Hz), 6.57 (1H, d, J=1.8 Hz),
3.90 (1H,
ddd, J=12.8, 12.8, 6.9 Hz), 1.94 (3H, s), 1.90-1.75 (2H, m), 1.69-1.50 (4H,
m), 1.46-1.32
10 (1H, m), 1.29-1.16 (1H, m)
Example 59
NC
N
0 N Me
CF3
4-15'-Methy1-2'-oxo-1'43-trifluoromethyl-phenyl)-2',3'-dihydro-l'H-
12,4'1biimidazoly1-1-yll-benzonitrile
15 Intermediate 11
NC
Br
4(2-Bromo-imidazol-1-y1)-benzonitrile
4-Imidazol-1-yl-benzonitrile (5.0 g, 29.6 mmol) was dissolved in dioxane and N-

bromosuccinimide (5.26 g, 29.6 mmol) was added. The solution was heated at 60
C for
20 2h. The solution was decanted from a gummy residue and evaporated to
give a yellow
solid. This was triturated with Et0Ac to give a cream solid which was further
purified by
chromatography using Et0Ac as eluent to afford Intermediate 11(0.79 g) as a
pale yellow

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
71
solid.
LCMS (Method 1) Rt = 2.44 min., m/z 248 and 250 (Br isotopes)
Intermediate 12
NC
0 N Me
CF3
4- f 2 ' -Ethoxy-5' -methyl-1 ' -(3-trifluo ro methyl-p h enyl)-1 'H- [2,4'l
biimidazoly1-
1-yll-benzonitrile
2-Dicyclohexylpho sphino -2 ',4 ',6'-triisopropylbiphenyl (19 mg, 0.04 mmol),
(2-
dicyclohexylpho sphino-2 ',4 ',6 '-triisopropy1-1,1 '-biphenyl)pal ladium(II)
dichloride (15.7
mg, 0.02 mmol), tetrahydroxydiboron (270 mg, 3 mmol) and potassium acetate
(294 mg, 3
mmol) were mixed in a nitrogen filled vial. A solution of 4-bromo-2-ethoxy-5-
methy1-1-
[3-(trifluoromethyl)phenyl]-1H-imidazole (Intermediate 4, 349 mg, 1 mmol) in
IMS (10
ml) was added and the solution heated at 80 C for 2h. A solution of K2CO3
(1.8M, 1.66m1)
was added followed by a suspension of 4-(2-bromo-imidazol-1-y1)-benzonitrile
(Intermediate 11, 248 mg, 1 mmol) in THF. Heating was continued overnight at
80 C.
After cooling, the mixture was filtered through celite, washing with Et0Ac,
and evaporated
to dryness. The residue was extracted into Et0Ac, decanted, dried over Na2SO4
filtered and
evaporated. Purification was performed by silica gel chromatography eluting
with 20% to
100% Et0Ac-cyclohexane. The third eluted component was the title compound (91
mg,
21%).
LCMS (Method 2) Rt = 2.73 min., m/z 438.3
4-1-5'-Methy1-2'-oxo-1'-(3-trifluoromethyl-phenyl)-2',3'-dihydro-l'H-
12,4'lbiimidazoly1-1-yll-benzonitrile (Example 59)
4- [2'-Ethoxy-5'-methy1-1'-(3-trifluoromethyl-pheny1)-1'H- [2,41] biimidazo
ly1-1 -yl] -

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
72
benzonitrile (Intermediate 12, 170 mg, 0.39 mmol), acetone (5 ml) and 1M HC1
(3 ml)
were heated together at 60 C for 3.5 h. Concentrated HC1 (0.5 ml) was added
and heating
continued overnight. The mixture was cooled and most of the acetone removed by

evaporation. The solution was basified with aqueous NaHCO3 and the white solid
title
compound filtered off and dried at 50 C. Yield 150 mg (94%).
LCMS (Method 3): Rt = 3.61 min, rniz 410.1 [M+H]+
1H NMR (400 MHz, d6-DMS0): 8 10.47 (1H, s), 8.05-7.99 (2H, m), 7.79-7.73 (3H,
m), 7.72 (1H, d, J=1.4 Hz), 7.68-7.63 (1H, m), 7.63-7.58 (2H, m), 7.28 (1H, d,
J=1.4 Hz),
1.70 (3H, s)
Example 60
NC
0
0,NANN
H
Me
C F3
1(4-Cyano-ph enyl)-5' -m ethyl-2' -oxo-1.' -(3-triflu orom ethyl-phenyl)-1',2
' -
dihydro-1H-12,4'lbiimidazoly1-3'-carboxylic acid cyclopentylamide
4- [5'-Methy1-2'-oxo-1'-(3 -trifluoromethyl-phenyl)-2',3 '-dihydro-1 'H-
[2,41biimidazoly1-1-y1]-benzonitrile (Example 59, 108 mg, 0.26 mmol) was
suspended in
dry DCM (5 ml) and DIPEA (1300, 3 eq.) was added followed by cyclopentyl
isocyanate
(90 p1, 88 mg, 0.79 mmol). The mixture was heated and stirred at 45 C for 24
h. The solvent
was removed in vacuo and the residue purified by chromatography eluting with
Et0Ac,
thereby yielding the title compound (100 mg, 73%).
LCMS (Method 3): Rt = 4.55 min, m/z 521.2 [M+H]+
1H NMR (400 MHz, d6-DMS0): 8 8.13 (1H, d, J=6.8 Hz), 8.02-7.98 (1H, m), 7.98-
7.93 (2H, m), 7.91-7.77 (3H, m), 7.72 (1H, d, J=1.4 Hz), 7.57-7.51 (2H, m),
7.20 (1H, d,
J=1.4 Hz), 3.75-3.65 (1H, m), 1.90 (3H, s), 1.75-1.57 (2H, m), 1.56-1.40 (4H,
m), 1.35-

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
73
0.94 (2H, m)
The following compounds were prepared by using an analogous procedure to that
in Example 60.
Table 7
Ex Structure 1H NMR LC-MS
Method
3
61 NC
'11NMR (400 MHz, Rt = 4.37 min,
CDC13): 6 8.51 (1H, d, m/z = 507.3
o 1`1.1
J=7.5 Hz), 7.78-7.70 [M+H]+
(311, m), 7.67 (111, t,
u N me J=7.9 Hz), 7.57-7.55
CF3 (111, m), 7.50-7.42 (311,
m), 7.33 (1H, d, J=1.3
1-(4-Cyano-phenyl)-5'-methyl-T- Hz), 7.27 (1H, d, J=1.3
oxo-1'-(3-trifluoromethyl-pheny1)- Hz), 4.15 (1H, tdt, J=8.0,
8.0, 8.1 Hz), 2.32-2.19
3'-carboxylic acid cyclobutylamide (2H, m), 1.87 (311, s),
1.94-1.75 (2H, m), 1.75-
1.58 (2H, m)
62 NC
NMR (400 MHz, d6- Rt = 4.05 min,
DMS0): 8 8.84(111, t, m/z = 621.3
o
J=6 Hz), 8.01 (111, m), [M+H]+
N1N N 7.93-7.80 (7H, m), 7.70
Ho.d,N¨Ime
b (111, d, J=1.5 Hz), 7.54-
CF, 7.51 (2H, m), 7.36-7.29
(2H, m), 7.20 (1H, d,
1-(4-Cyano-phenyl)-5'-methyl-2'- J=1.5 Hz), 4.28 (2H, d,
oxo-1'-(3-trifluoromethyl-pheny1)- J=5.3 Hz), 3.21 (3H, s),
1.89 (311, s)
3'-carboxylic acid 4-
methanesulfonyl-benzylamide
Example
NC
-N
0 N
H
OdNi Me
CF3

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
74
5-[2-(4-Cyano-pheny1)-2H-[1,2,41triazol-3-y11-4-methyl-2-oxo-3-(3-
trifluoromethyl-pheny1)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide
Intermediate 13
NC
-N
Br
4-(5-B romo- [1,2,41 triazo 1-1-y1)-benzonitrile
441,2,4]Triazol-1-yl-benzonitrile (1.7 g, 10 mmol) and N-bromosuccinimide
(1.78
g, 10 mmol) were heated together in dioxane (50 ml) at 60 C for 3h.The mixture
was
filtered and the filtrate evaporated and the resulting solid was purified by
chromatography
to give Intermediate 13 as a white solid (0.6 g, 25%).
LCMS (Method 3) Rt = 2.61 min., m/z 249 and 251 (Br isotopes)
Intermediate 14
NC = N-N
0 N Me
CF3
445-1-2-Ethoxy-5-methy1-1-(3-trifluoromethyl-pheny1)-1H-imidazol-4-y11-
11,2,41triazol-1-y11-benzonitrile
4-Bromo-2-ethoxy-5-methyl-143-(trifluoromethyl)phenyl] -1H-imidazole
(Intermediate 4, 349 mg, 1 mmol), 2-dicyclohexylphosphino-2',4',6'-
triisopropylbiphenyl
(19 mg, 0.04 mmol), (2-dicyclohexylpho sphino -2 ',4',6 '-trii
sopropyl-1,1 '-
biphenyl)palladium(II) dichloride (15.7 mg, 0.02 mmol), tetrahydroxydiboron
(270 mg, 3
mmol) and potassium acetate (294 mg, 3 mmol) and IMS (10 ml) were sealed in
each of 2
vials under N2 and heated at 80 C for 1.5 h, then 4-(5-bromo-[1,2,4]triazol-1-
y1)-
benzonitrile (Intermediate 13, 249 mg, 1 mmol) and 1.8M K2CO3 (1.66 ml) were
added to
each vial and heating continued for a further 1 h. The reactions were
combined, filtered and

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
evaporated and the residue triturated with DCM to remove more inorganic
solids. The
solution was chromatographed on silica (0% to 50% Et0Ac-DCM) to provide
Intermediate
14 (100 mg, 11.5%).
LCMS (Method U2) Rt = 1.55 min., m/z 439.2
5 Intermediate 15
NC
N Me
C F3
4-{5-1-5-Methyl-2-oxo-1-(3-trifluoromethyl-phenyl)-2,3-dihydro-1H-imidazol-
4-y11-1-1,2,41triazol-1-y1}-benzonitrile
4- {5- [2-Ethoxy-5 -methyl-1 -(3 -trifluoromethyl-pheny1)-1H-imidazol-4-y1]-
10 [1,2,4]triazol-1-y1}-benzonitrile (Intermediate 14, 130 mg, 0.30 mmol),
acetone (4 ml) and
5M HC1 (3 ml) were heated together at 60 C for 2 h and allowed to cool. The
acetone was
removed in vacuo and the aqueous solution basified with NaHCO3 solution. The
white solid
Intermediate 15 was filtered off, washed with water and dried (79 mg, 66%).
LCMS (Method U2) Rt = 1.33 min., m/z 411.1
15 5-[2-(4-Cyano-phenyl)-2H-1-1,2,41triazol-3-y11-4-methyl-2-oxo-3-(3-
trifluoromethyl-pheny1)-2,3-dihydro-imidazole-1-carboxylic acid
cyclopentylamide
(Example 63)
4- {5- [5-Methy1-2-oxo-1-(3-trifluoromethyl-pheny1)-2,3-dihydro-1H-imidazol-4-
y1]-[1,2,4]triazol-1-yll-benzonitrile (Intermediate 15, 79 mg, 0.192 mmol),
cyclopentyl
20 isocyanate (58 jtl, 0.58 mmol) and DIPEA (101 p1, 0.58 mmol) were
stirred in DCM (4 ml)
at 45 C overnight. Solvent was evaporated and the product was purified by
silica gel
chromatography (50% to 100% Et0Ac-cyclohexane) to give title compound as a
white
solid (73 mg, 73%).

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
76
LCMS (Method 3): Rt = 5.16 mm, m/z 522.3 [M+H]+
1H NMR (400 MHz, CDC13): 8 8.27 (1H, d, J=6.8 Hz), 8.19 (1H, s), 7.81-7.74
(3H,
m), 7.70 (1H, tm, J=8 Hz), 7.67-7.63 (2H, m), 7.61 (1H, m), 7.52 (1H, dm, J=8
Hz), 3.98-
3.87 (1H, m), 1.93 (3H, s), 1.91-1.73 (2H, m), 1.69-1.52 (5H, m), 1.47-1.35
(111, m)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
77
Biological Assay
Compounds of this invention were tested for potency in a human neutrophil
elastase
(HNE) enzyme activity assay.
HNE Enzyme Assay
Assays were performed in 96-well plates in a total assay volume of 100 L. The
final
concentration of elastase enzyme (human leukocyte elastase, Sigma E8140) was
0.00072
U/mL. The peptide substrate (Me0Suc-Ala-Ala-Pro-Val-AMC, Calbiochem #324740)
was
used at a final concentration of 100 M. The final concentration of DMSO was
1% in the assay
buffer (0.05M Tris.HC1, 0.1M NaC1, 0.1M CaC12, 0.0005% brij-35, pH 7.5). The
enzymatic
reaction was started by addition of the enzyme and incubated at 25 C for 30
minutes. After
incubation, the reaction was stopped by addition of soybean trypsin inhibitor
(Sigma T9003) at
a fmal concentration of 50 jig/well. Fluorescence was measured using a
Molecular Devices
fluorescence plate reader using 380 nm excitation and 460 nm emission
wavelengths.
A dose response to each compound was performed and the effect of compound in
each experiment was expressed as a percentage inhibition of the control enzyme
fluorescence. Dose response curves were plotted and compound potency (ICso)
was
determined. Compounds were tested in at least two separate experiments. ICso
values for
tested Examples, representative of the invention, are shown in Table 8:
Table 8
Example HNE inhibition
1 ++
2 +++
3 +++
4 ++++
5 +++
6 ++++
7 -F-
8 ++++
(continued)

CA 02970664 2017-06-12
WO 2016/096638
PCT/EP2015/079371
78
9 +++
10 -H-++
11 ++++
12 +++
13 ++++
14 ++++
15 ++++
16 ++++
17 ++++
18 ++++
19 ++++
20 ++
21 ++
22 ++
23 ++
24 +++
25 +++
26 ii
27 ++++
28 +++
29 +++
30 +++
31 +++
32 ++++
33 +++
34 ++++
35 +++
36 +++
37 +++
38 +++
39 ++
40 +++
(continued)

CA 02970664 2017-06-12
WO 2016/096638 PCT/EP2015/079371
79
41 -H-++
42 ++++
43 ++++
44 ++++
45 ++++
46 ++++
47 ++++
48 ++++
49 +++
50 +++
51 ++++
52 +++
53 ++++
54 +++
56 +++
57 ++
58 ++++
59 ++
++++
61 ++++
62 ++++
63 ++++
In the table above, HNE enzyme inhibition (IC50 values) are indicated as
follows: >
50 nM `+`; 10-50 nM `++'; 1-10 nM `+++'; <1 nM `++++'.

Dessin représentatif

Désolé, le dessin représentatatif concernant le document de brevet no 2970664 est introuvable.

États administratifs

Pour une meilleure compréhension de l'état de la demande ou brevet qui figure sur cette page, la rubrique Mise en garde , et les descriptions de Brevet , États administratifs , Taxes périodiques et Historique des paiements devraient être consultées.

États administratifs

Titre Date
Date de délivrance prévu Non disponible
(86) Date de dépôt PCT 2015-12-11
(87) Date de publication PCT 2016-06-23
(85) Entrée nationale 2017-06-12
Demande morte 2019-12-11

Historique d'abandonnement

Date d'abandonnement Raison Reinstatement Date
2018-12-11 Taxe périodique sur la demande impayée

Historique des paiements

Type de taxes Anniversaire Échéance Montant payé Date payée
Le dépôt d'une demande de brevet 400,00 $ 2017-06-12
Taxe de maintien en état - Demande - nouvelle loi 2 2017-12-11 100,00 $ 2017-06-12
Titulaires au dossier

Les titulaires actuels et antérieures au dossier sont affichés en ordre alphabétique.

Titulaires actuels au dossier
CHIESI FARMACEUTICI S.P.A.
Titulaires antérieures au dossier
S.O.
Les propriétaires antérieurs qui ne figurent pas dans la liste des « Propriétaires au dossier » apparaîtront dans d'autres documents au dossier.
Documents

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :



Pour visualiser une image, cliquer sur un lien dans la colonne description du document. Pour télécharger l'image (les images), cliquer l'une ou plusieurs cases à cocher dans la première colonne et ensuite cliquer sur le bouton "Télécharger sélection en format PDF (archive Zip)" ou le bouton "Télécharger sélection (en un fichier PDF fusionné)".

Liste des documents de brevet publiés et non publiés sur la BDBC .

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.


Description du
Document 
Date
(yyyy-mm-dd) 
Nombre de pages   Taille de l'image (Ko) 
Lettre du bureau 2020-01-03 1 203
Abrégé 2017-06-12 1 51
Revendications 2017-06-12 9 354
Description 2017-06-12 79 2 978
Rapport de recherche internationale 2017-06-12 3 88
Demande d'entrée en phase nationale 2017-06-12 5 137
Poursuite - Modification 2017-06-12 11 389
Page couverture 2017-08-21 1 26